Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Cell ; 170(5): 973-985.e10, 2017 Aug 24.
Article in English | MEDLINE | ID: mdl-28841420

ABSTRACT

Mycobacterium leprae causes leprosy and is unique among mycobacterial diseases in producing peripheral neuropathy. This debilitating morbidity is attributed to axon demyelination resulting from direct interaction of the M. leprae-specific phenolic glycolipid 1 (PGL-1) with myelinating glia and their subsequent infection. Here, we use transparent zebrafish larvae to visualize the earliest events of M. leprae-induced nerve damage. We find that demyelination and axonal damage are not directly initiated by M. leprae but by infected macrophages that patrol axons; demyelination occurs in areas of intimate contact. PGL-1 confers this neurotoxic response on macrophages: macrophages infected with M. marinum-expressing PGL-1 also damage axons. PGL-1 induces nitric oxide synthase in infected macrophages, and the resultant increase in reactive nitrogen species damages axons by injuring their mitochondria and inducing demyelination. Our findings implicate the response of innate macrophages to M. leprae PGL-1 in initiating nerve damage in leprosy.


Subject(s)
Antigens, Bacterial/metabolism , Disease Models, Animal , Glycolipids/metabolism , Leprosy/microbiology , Leprosy/pathology , Macrophages/immunology , Mycobacterium leprae/physiology , Animals , Axons/metabolism , Axons/pathology , Demyelinating Diseases , Larva/growth & development , Leprosy/immunology , Mycobacterium marinum/metabolism , Myelin Sheath/chemistry , Myelin Sheath/metabolism , Myelin Sheath/ultrastructure , Neuroglia/metabolism , Neuroglia/pathology , Nitric Oxide/metabolism , Zebrafish
2.
PLoS Pathog ; 13(7): e1006496, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28704551

ABSTRACT

Successful host defense against pathogens requires innate immune recognition of the correct pathogen associated molecular patterns (PAMPs) by pathogen recognition receptors (PRRs) to trigger the appropriate gene program tailored to the pathogen. While many PRR pathways contribute to the innate immune response to specific pathogens, the relative importance of each pathway for the complete transcriptional program elicited has not been examined in detail. Herein, we used RNA-sequencing with wildtype and mutant macrophages to delineate the innate immune pathways contributing to the early transcriptional response to Staphylococcus aureus, a ubiquitous microorganism that can activate a wide variety of PRRs. Unexpectedly, two PRR pathways-the Toll-like receptor (TLR) and Stimulator of Interferon Gene (STING) pathways-were identified as dominant regulators of approximately 95% of the genes that were potently induced within the first four hours of macrophage infection with live S. aureus. TLR signaling predominantly activated a pro-inflammatory program while STING signaling activated an antiviral/type I interferon response with live but not killed S. aureus. This STING response was largely dependent on the cytosolic DNA sensor cyclic guanosine-adenosine synthase (cGAS). Using a cutaneous infection model, we found that the TLR and STING pathways played opposite roles in host defense to S. aureus. TLR signaling was required for host defense, with its absence reducing interleukin (IL)-1ß production and neutrophil recruitment, resulting in increased bacterial growth. In contrast, absence of STING signaling had the opposite effect, enhancing the ability to restrict the infection. These results provide novel insights into the complex interplay of innate immune signaling pathways triggered by S. aureus and uncover opposing roles of TLR and STING in cutaneous host defense to S. aureus.


Subject(s)
Cytosol/immunology , Membrane Proteins/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/physiology , Toll-Like Receptors/immunology , Animals , Cytosol/microbiology , DNA , DNA, Bacterial/genetics , DNA, Bacterial/immunology , Female , Humans , Immunity, Innate , Interferon Type I/genetics , Interferon Type I/immunology , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Nucleotidyltransferases/genetics , Nucleotidyltransferases/immunology , Signal Transduction , Staphylococcal Infections/genetics , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Toll-Like Receptors/genetics
3.
PLoS Pathog ; 12(6): e1005705, 2016 06.
Article in English | MEDLINE | ID: mdl-27355424

ABSTRACT

Triggering antimicrobial mechanisms in macrophages infected with intracellular pathogens, such as mycobacteria, is critical to host defense against the infection. To uncover the unique and shared antimicrobial networks induced by the innate and adaptive immune systems, gene expression profiles generated by RNA sequencing (RNAseq) from human monocyte-derived macrophages (MDMs) activated with TLR2/1 ligand (TLR2/1L) or IFN-γ were analyzed. Weighed gene correlation network analysis identified modules of genes strongly correlated with TLR2/1L or IFN-γ that were linked by the "defense response" gene ontology term. The common TLR2/1L and IFN-γ inducible human macrophage host defense network contained 16 antimicrobial response genes, including S100A12, which was one of the most highly induced genes by TLR2/1L. There is limited information on the role of S100A12 in infectious disease, leading us to test the hypothesis that S100A12 contributes to host defense against mycobacterial infection in humans. We show that S100A12 is sufficient to directly kill Mycobacterium tuberculosis and Mycobacterium leprae. We also demonstrate that S100A12 is required for TLR2/1L and IFN-γ induced antimicrobial activity against M. leprae in infected macrophages. At the site of disease in leprosy, we found that S100A12 was more strongly expressed in skin lesions from tuberculoid leprosy (T-lep), the self-limiting form of the disease, compared to lepromatous leprosy (L-lep), the progressive form of the disease. These data suggest that S100A12 is part of an innate and adaptive inducible antimicrobial network that contributes to host defense against mycobacteria in infected macrophages.


Subject(s)
Leprosy/immunology , Macrophages/immunology , S100A12 Protein/immunology , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , Macrophages/microbiology , Mycobacterium Infections/immunology , Mycobacterium leprae/immunology , Mycobacterium tuberculosis/immunology , Real-Time Polymerase Chain Reaction , Transcriptome
4.
J Immunol ; 194(3): 1169-77, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25548220

ABSTRACT

Current evidence suggests that neonatal immunity is functionally distinct from adults. Although TLR signaling through the adaptor protein, MyD88, has been shown to be critical for survival to sepsis in adults, little is known about the role of MyD88 or TRIF in neonatal sepsis. We demonstrate that TRIF(-/-) but not MyD88(-/-) neonates are highly susceptible to Escherichia coli peritonitis and bacteremia. This was associated with decreased innate immune recruitment and function. Importantly, we found that the reverse was true in adults that MyD88(-/-) but not TRIF(-/-) or wild-type adults are susceptible to E. coli peritonitis and bacteremia. In addition, we demonstrate that TRIF but not MyD88 signaling is critical for the TLR4 protective adjuvant effect we have previously demonstrated. These data suggest a differential requirement for the survival of neonates versus adults to Gram-negative infection, and that modulation of TRIF in neonates can be used to augment survival to neonatal sepsis.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Gram-Negative Bacterial Infections/genetics , Gram-Negative Bacterial Infections/immunology , Immunity, Innate , Sepsis/genetics , Sepsis/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Animals, Newborn , Chemokine CXCL10/metabolism , Chemokines/biosynthesis , Cytokines/biosynthesis , Disease Models, Animal , Disease Susceptibility/immunology , Escherichia coli/immunology , Female , Genetic Predisposition to Disease , Gram-Negative Bacterial Infections/metabolism , Gram-Negative Bacterial Infections/microbiology , Gram-Negative Bacterial Infections/mortality , Granulocytes/immunology , Granulocytes/metabolism , Interferon Type I/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Knockout , Neutrophils/immunology , Neutrophils/metabolism , Phagocytosis/genetics , Phagocytosis/immunology , Reactive Oxygen Species/metabolism , Sepsis/metabolism , Sepsis/microbiology , Sepsis/mortality , Toll-Like Receptors/metabolism
5.
J Immunol ; 192(12): 6111-9, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24829407

ABSTRACT

Cancer progression is associated with inflammation, increased metabolic demand, infection, cachexia, and eventually death. Myeloid-derived suppressor cells (MDSCs) commonly expand during cancer and are associated with adaptive immune suppression and inflammatory metabolite production. We propose that cancer-induced cachexia is driven at least in part by the expansion of MDSCs. MDSC expansion in 4T1 mammary carcinoma-bearing hosts is associated with induction of a hepatic acute-phase protein response and altered host energy and fat metabolism, and eventually reduced survival to polymicrobial sepsis and endotoxemia. Similar results are also seen in mice bearing a Lewis lung carcinoma and a C26 colon adenocarcinoma. However, a similar cachexia response is not seen with equivalent growth of the 66C4 subclone of 4T1, in which MDSC expansion does not occur. Importantly, reducing MDSC numbers in 4T1-bearing animals can ameliorate some of these late responses and reduce susceptibility to inflammation-induced organ injury and death. In addition, administering MDSCs from both tumor- and nontumor-bearing mice can produce an acute-phase response. Thus, we propose a previously undescribed mechanism for the development of cancer cachexia, whereby progressive MDSC expansion contributes to changes in host protein and energy metabolism and reduced resistance to infection.


Subject(s)
Cachexia/immunology , Immune Tolerance , Myeloid Cells/immunology , Neoplasms, Experimental/immunology , Animals , Cachexia/etiology , Cell Line, Tumor , Female , Mice , Mice, Inbred BALB C , Myeloid Cells/pathology , Neoplasms, Experimental/pathology
6.
J Immunol ; 190(8): 3916-27, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23509349

ABSTRACT

Although ectopic lymphoid tissue formation is associated with many autoimmune diseases, it is unclear whether it serves a functional role in autoimmune responses. 2,6,10,14-Tetramethylpentadecane causes chronic peritoneal inflammation and lupus-like disease with autoantibody production and ectopic lymphoid tissue (lipogranuloma) formation. A novel transplantation model was used to show that transplanted lipogranulomas retain their lymphoid structure over a prolonged period in the absence of chronic peritoneal inflammation. Recipients of transplanted lipogranulomas produced anti-U1A autoantibodies derived exclusively from the donor, despite nearly complete repopulation of the transplanted lipogranulomas by host lymphocytes. The presence of ectopic lymphoid tissue alone was insufficient, as an anti-U1A response was not generated by the host in the absence of ongoing peritoneal inflammation. Donor-derived anti-U1A autoantibodies were produced for up to 2 mo by plasma cells/plasmablasts recruited to the ectopic lymphoid tissue by CXCR4. Although CD4(+) T cells were not required for autoantibody production from the transplanted lipogranulomas, de novo generation of anti-U1A plasma cells/plasmablasts was reduced following T cell depletion. Significantly, a population of memory B cells was identified in the bone marrow and spleen that did not produce anti-U1A autoantibodies unless stimulated by LPS to undergo terminal differentiation. We conclude that 2,6,10,14-tetramethylpentadecane promotes the T cell-dependent development of class-switched, autoreactive memory B cells and plasma cells/plasmablasts. The latter home to ectopic lymphoid tissue and continue to produce autoantibodies after transplantation and in the absence of peritoneal inflammation. However, peritoneal inflammation appears necessary to generate autoreactive B cells de novo.


Subject(s)
Autoantibodies/biosynthesis , B-Lymphocyte Subsets/immunology , Bone Marrow Cells/immunology , Immunologic Memory , Lymphoid Tissue/immunology , Plasma Cells/immunology , Ribonucleoproteins, Small Nuclear/immunology , Animals , Autoantibodies/metabolism , B-Lymphocyte Subsets/cytology , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Choristoma/immunology , Female , Granuloma/blood , Granuloma/immunology , Granuloma/pathology , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Plasma Cells/metabolism
7.
J Exp Med ; 204(6): 1463-74, 2007 Jun 11.
Article in English | MEDLINE | ID: mdl-17548519

ABSTRACT

Polymicrobial sepsis alters the adaptive immune response and induces T cell suppression and Th2 immune polarization. We identify a GR-1(+)CD11b(+) population whose numbers dramatically increase and remain elevated in the spleen, lymph nodes, and bone marrow during polymicrobial sepsis. Phenotypically, these cells are heterogeneous, immature, predominantly myeloid progenitors that express interleukin 10 and several other cytokines and chemokines. Splenic GR-1(+) cells effectively suppress antigen-specific CD8(+) T cell interferon (IFN) gamma production but only modestly suppress antigen-specific and nonspecific CD4(+) T cell proliferation. GR-1(+) cell depletion in vivo prevents both the sepsis-induced augmentation of Th2 cell-dependent and depression of Th1 cell-dependent antibody production. Signaling through MyD88, but not Toll-like receptor 4, TIR domain-containing adaptor-inducing IFN-beta, or the IFN-alpha/beta receptor, is required for complete GR-1(+)CD11b(+) expansion. GR-1(+)CD11b(+) cells contribute to sepsis-induced T cell suppression and preferential Th2 polarization.


Subject(s)
CD11b Antigen/metabolism , Immune Tolerance/immunology , Myeloid Differentiation Factor 88/metabolism , Myeloid Progenitor Cells/immunology , Receptors, Chemokine/metabolism , Sepsis/immunology , Th2 Cells/immunology , Animals , Cell Proliferation , Colony-Forming Units Assay , Cytokines/metabolism , Flow Cytometry , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Mice , Mice, Inbred C57BL , Myeloid Progenitor Cells/metabolism , Signal Transduction/immunology
8.
J Immunol ; 187(2): 911-8, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21690321

ABSTRACT

Neutrophils are essential for successful host eradication of bacterial pathogens and for survival to polymicrobial sepsis. During inflammation, the bone marrow provides a large reserve of neutrophils that are released into the peripheral circulation where they traverse to sites of infection. Although neutrophils are essential for survival, few studies have investigated the mechanisms responsible for neutrophil mobilization from the bone marrow during polymicrobial sepsis. Using a cecal ligation and puncture model of polymicrobial sepsis, we demonstrated that neutrophil mobilization from the bone marrow is not dependent on TLR4, MyD88, TRIF, IFNARα/ß, or CXCR2 pathway signaling during sepsis. In contrast, we observed that bone marrow CXCL12 mRNA abundance and specific CXCL12 levels are sharply reduced, whereas splenic CXCR4 mRNA and cell surface expression are increased during sepsis. Blocking CXCL12 activity significantly reduced blood neutrophilia by inhibiting bone marrow release of granulocytes during sepsis. However, CXCL12 inhibition had no impact on the expansion of bone marrow neutrophil precursors and hematopoietic progenitors. Bone marrow neutrophil retention by CXCL12 blockade prevented blood neutrophilia, inhibited peritoneal neutrophil accumulation, allowed significant peritoneal bacterial invasion, and increased polymicrobial sepsis mortality. We concluded that changes in the pattern of CXCL12 signaling during sepsis are essential for neutrophil bone marrow mobilization and host survival but have little impact on bone marrow granulopoiesis.


Subject(s)
Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Chemokine CXCL12/physiology , Neutrophil Infiltration/immunology , Sepsis/immunology , Sepsis/microbiology , Signal Transduction/immunology , Acute Disease , Animals , Bone Marrow Cells/microbiology , Chemokine CXCL12/genetics , Mice , Mice, 129 Strain , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myelopoiesis/genetics , Myelopoiesis/immunology , Neutrophil Infiltration/genetics , Sepsis/mortality , Signal Transduction/genetics , Survival Analysis
9.
J Immunol ; 186(1): 195-202, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21106855

ABSTRACT

Sepsis, the systemic inflammatory response to microbial infection, induces changes in both innate and adaptive immunity that presumably lead to increased susceptibility to secondary infections, multiorgan failure, and death. Using a model of murine polymicrobial sepsis whose severity approximates human sepsis, we examined outcomes and defined requirements for survival after secondary Pseudomonas aeruginosa pneumonia or disseminated Listeria monocytogenes infection. We demonstrate that early after sepsis neutrophil numbers and function are decreased, whereas monocyte recruitment through the CCR2/MCP-1 pathway and function are enhanced. Consequently, lethality to Pseudomonas pneumonia is increased early but not late after induction of sepsis. In contrast, lethality to listeriosis, whose eradication is dependent upon monocyte/macrophage phagocytosis, is actually decreased both early and late after sepsis. Adaptive immunity plays little role in these secondary infectious responses. This study demonstrates that sepsis promotes selective early, impaired innate immune responses, primarily in neutrophils, that lead to a pathogen-specific, increased susceptibility to secondary infections.


Subject(s)
Bacteremia/immunology , Bacteremia/mortality , Immunity, Innate , Sepsis/immunology , Sepsis/mortality , Animals , Bacteremia/pathology , Cecum , Disease Models, Animal , Genetic Predisposition to Disease , Immunity, Innate/genetics , Ligation , Listeriosis/immunology , Listeriosis/mortality , Listeriosis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Neutrophils/pathology , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/mortality , Pneumonia, Bacterial/pathology , Pseudomonas Infections/immunology , Pseudomonas Infections/mortality , Pseudomonas Infections/pathology , Punctures , Sepsis/pathology , Time Factors
10.
Adv Sci (Weinh) ; 10(31): e2302248, 2023 11.
Article in English | MEDLINE | ID: mdl-37750461

ABSTRACT

New vaccine platforms that activate humoral immunity and generate neutralizing antibodies are required to combat emerging pathogens, including influenza virus. A slurry of antigen-loaded hydrogel microparticles that anneal to form a porous scaffold with high surface area for antigen uptake by infiltrating immune cells as the biomaterial degrades is demonstrated to enhance humoral immunity. Antigen-loaded-microgels elicited a robust cellular humoral immune response, with increased CD4+ T follicular helper (Tfh) cells and prolonged germinal center (GC) B cells comparable to the commonly used adjuvant, aluminum hydroxide (Alum). Increasing the weight fraction of polymer material led to increased material stiffness and antigen-specific antibody titers superior to Alum. Vaccinating mice with inactivated influenza virus loaded into this more highly cross-linked formulation elicited a strong antibody response and provided protection against a high dose viral challenge. By tuning physical and chemical properties, adjuvanticity can be enhanced leading to humoral immunity and protection against a pathogen, leveraging two different types of antigenic material: individual protein antigen and inactivated virus. The flexibility of the platform may enable design of new vaccines to enhance innate and adaptive immune cell programming to generate and tune high affinity antibodies, a promising approach to generate long-lasting immunity.


Subject(s)
Influenza Vaccines , Influenza, Human , Orthomyxoviridae Infections , Orthomyxoviridae , Animals , Mice , Humans , Immunity, Humoral , Porosity , Antibodies, Viral , Antigens
11.
J Immunol ; 184(5): 2247-51, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20130216

ABSTRACT

Bone marrow (BM) hematopoietic stem and progenitor cells (HSPCs) can be activated by type I IFNs, TLR agonists, viruses, and bacteria to increase hematopoiesis. In this study, we report that endotoxin treatment in vivo induces TLR4, MyD88, and Toll/IL-1 resistance domain-containing adaptor-inducing IFN-beta (TRIF)-dependent expansion of BM HSPCs. Bacterial infection by Staphylococcus aureus or cecal ligation and puncture also induces HSPC expansion, but MyD88, TRIF, type I IFN, cytokine, PG, or oxidative stress pathways are not required for their expansion. S. aureus-induced HSPC expansion in MyD88(-/-)TRIF(-/-) mice is also normal, but is associated with BM remodeling as granulocyte stores are released peripherally. Importantly, reduction in BM cellularity alone can reproduce HSPC expansion. These data show in vivo HSPC responses to bacterial infection are complex and not absolutely dependent upon key inflammatory signaling pathways.


Subject(s)
Bacterial Infections/immunology , Hematopoietic Stem Cells/immunology , Signal Transduction/immunology , Stem Cells/immunology , Toll-Like Receptor 4/immunology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Animals , Bacterial Infections/etiology , Bacterial Infections/microbiology , Bone Marrow/immunology , Bone Marrow/metabolism , Bone Marrow/microbiology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cecum/surgery , Cell Proliferation/drug effects , Cells, Cultured , Flow Cytometry , Hematopoietic Stem Cells/cytology , Interleukin-6/genetics , Interleukin-6/immunology , Ligation/adverse effects , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/immunology , Signal Transduction/drug effects , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Stem Cells/cytology , Toll-Like Receptor 4/genetics
12.
Infect Immun ; 79(7): 2746-54, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21518789

ABSTRACT

Previous studies have suggested that neonates rely heavily on innate immunity for their antimicrobial response to bacterial infections. However, the innate immune response by neonates to bacterial infection remains poorly characterized. Here, we show that in a murine model of neonatal polymicrobial sepsis, CXC ligand 10 (CXCL10) concentrations increase in the blood and peritoneum concordant with the peritoneal recruitment of granulocytes and macrophages. Additionally, CXC receptor 3 (CXCR3) expression on elicited peritoneal macrophages and granulocytes increases following sepsis. Blockade of CXCL10 worsens not only recruitment and phagocytic function of peritoneal granulocytes and macrophages but also survival. Deletion of CXCR3 also significantly increases mortality to a septic challenge. Finally, we demonstrate that the protective adjuvant effect of pretreatment with a Toll-like receptor 4 agonist to neonatal sepsis is dependent on an endogenous CXCL10 response and that pretreatment of neonates with CXCL10 can also significantly improve macrophage and granulocyte function and modestly improve outcome to polymicrobial sepsis. Together, these data suggest a critical role for CXCL10 signaling during neonatal sepsis.


Subject(s)
Bacterial Infections/immunology , Chemokine CXCL10/immunology , Receptors, CXCR3/metabolism , Sepsis/immunology , Signal Transduction , Animals , Animals, Newborn , Bacterial Infections/metabolism , Chemokine CXCL10/antagonists & inhibitors , Chemokine CXCL10/blood , Female , Flow Cytometry , Granulocytes/immunology , Granulocytes/metabolism , Immunity, Innate , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis , Receptors, CXCR3/blood , Receptors, CXCR3/deficiency , Receptors, CXCR3/genetics , Sepsis/metabolism , Sepsis/microbiology , Toll-Like Receptor 4/immunology
13.
Lab Invest ; 91(10): 1540-50, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21808234

ABSTRACT

Diffuse alveolar hemorrhage is an uncommon, yet often fatal, complication of systemic lupus erythematosus (SLE). Advances in the treatment of alveolar hemorrhage have been hampered because of the heterogeneity of clinical findings and the lack of suitable animal models. A single intraperitoneal injection of pristane induces a lupus-like syndrome characterized by lupus-related autoantibodies and glomerulonephritis in non-autoimmune-prone strains of mice. In addition, C57BL/6 (B6) mice frequently develop alveolar hemorrhage within a few weeks of pristane injection. Immunopathogenesis of pristane-induced alveolar hemorrhage was investigated in the present study. Early (2-4 weeks after injection) mortality due to hemorrhage was unique to C57BL/6 and C57BL/10 strains of mice. Recruitment of the macrophages and neutrophils preceded the hemorrhage by several days, and hemorrhage started 3-7 days after pristane injection in some mice, peaked at 2 weeks (84% in B6) and then resolved by 4 weeks in a majority of mice. Alveolar hemorrhage was independent of MyD88 (myeloid differentiation factor 88), or TLR7 pathways, in contrast to autoantibody production and glomerulonephritis, and was also independent of FcγR or Fas. Rag1(-/-) mice had a reduced prevalence of alveolar hemorrhage compared with B6 (P=0.01) congenics. However, T-cell receptor-deficient mice developed alveolar hemorrhage at a rate comparable to wild-type controls, whereas B6 Igµ(-/-) mice surprisingly had a strikingly reduced prevalence (7% vs 84% in B6, P<0.0001). Reconstitution of B6 Igµ(-/-) mice with wild-type B cells increased the prevalence to 50% (P=0.028). Pristane-induced alveolar hemorrhage is a useful model to study the pathogenesis and develop new therapy for this underappreciated and often life-threatening complication of SLE.


Subject(s)
B-Lymphocytes , Hemorrhage/chemically induced , Lung Diseases/chemically induced , Pulmonary Alveoli , Terpenes , Animals , B-Lymphocytes/pathology , Cell Line , Hemorrhage/pathology , Homeodomain Proteins/genetics , Humans , Intercellular Signaling Peptides and Proteins , Lung/drug effects , Lung/pathology , Lung Diseases/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Species Specificity , Substrate Specificity
14.
Lab Invest ; 91(12): 1787-95, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21931299

ABSTRACT

Gene expression analysis can be a powerful tool in predicting patient outcomes and identifying patients who may benefit from targeted therapies. However, isolating human blood polymorphonuclear cells (PMNs) for genomic analysis has been challenging. We used a novel microfluidic technique that isolates PMNs by capturing CD66b(+) cells and compared it with dextran-Ficoll gradient isolation. We also used microfluidic isolation techniques for blood and bronchoalveolar lavage (BAL) samples of patients with acute respiratory distress syndrome (ARDS) to evaluate PMN genomic alterations secondary to pulmonary sequestration. PMNs obtained from ex vivo lipopolysaccharide (LPS)-stimulated or -unstimulated whole blood from five healthy volunteers were isolated by either dextran-Ficoll gradient, microfluidics capture, or a combination of the two techniques. Blood and BAL fluid PMNs were also isolated using microfluidics from seven hospitalized patients with ARDS. Gene expression was inferred from extracted RNA using Affymetrix U133 Plus 2.0 GeneChips. All methods of PMN isolation produced similar quantities of high-quality RNA, when adjusted for recovered cell number. Unsupervised analysis and hierarchical clustering indicated that LPS stimulation was the primary factor affecting gene expression patterns among all ex vivo samples. Patterns of gene expression from blood and BAL PMNs differed significantly from each other in the patients with ARDS. Isolation of PMNs by microfluidics can be applied to both blood and BAL specimens from critically ill, hospitalized patients. Unique genomic expression patterns are obtained from the blood and BAL fluid of critically ill patients with ARDS, and these differ significantly from genomic patterns seen after ex vivo LPS stimulation.


Subject(s)
Acute Lung Injury/pathology , Antigens, CD/analysis , Bronchoalveolar Lavage Fluid/cytology , Cell Adhesion Molecules/analysis , Neutrophils/pathology , Respiratory Distress Syndrome/pathology , Acute Lung Injury/blood , Case-Control Studies , Centrifugation, Density Gradient , GPI-Linked Proteins/analysis , Gene Expression Profiling , Humans , Microfluidic Analytical Techniques , RNA/isolation & purification , Respiratory Distress Syndrome/blood
15.
Mol Med ; 17(3-4): 281-92, 2011.
Article in English | MEDLINE | ID: mdl-21085745

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are a heterogenous population of immature myeloid cells whose numbers dramatically increase in chronic and acute inflammatory diseases, including cancer, autoimmune disease, trauma, burns and sepsis. Studied originally in cancer, these cells are potently immunosuppressive, particularly in their ability to suppress antigen-specific CD8(+) and CD4(+) T-cell activation through multiple mechanisms, including depletion of extracellular arginine, nitrosylation of regulatory proteins, and secretion of interleukin 10, prostaglandins and other immunosuppressive mediators. However, additional properties of these cells, including increased reactive oxygen species and inflammatory cytokine production, as well as their universal expansion in nearly all inflammatory conditions, suggest that MDSCs may be more of a normal component of the inflammatory response ("emergency myelopoiesis") than simply a pathological response to a growing tumor. Recent evocative data even suggest that the expansion of MDSCs in acute inflammatory processes, such as burns and sepsis, plays a beneficial role in the host by increasing immune surveillance and innate immune responses. Although clinical efforts are currently underway to suppress MDSC numbers and function in cancer to improve antineoplastic responses, such approaches may not be desirable or beneficial in other clinical conditions in which immune surveillance and antimicrobial activities are required.


Subject(s)
Lymphocyte Activation/immunology , Myeloid Cells/immunology , Sepsis/immunology , Wounds and Injuries/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Humans , Models, Immunological
16.
J Immunol ; 182(7): 4226-36, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19299721

ABSTRACT

Intraperitoneal exposure of nonautoimmune mice to 2,6,10,14-tetramethylpentadecane (TMPD) causes lupus and the formation of ectopic lymphoid tissue. Although associated with humoral autoimmunity, it is not known whether Ab responses develop within ectopic lymphoid tissue or if B cells only secondarily migrate there. We show that ectopic lymphoid tissue induced by TMPD not only resembles secondary lymphoid tissue morphologically, but it also displays characteristics of germinal center reactions. Proliferating T and B lymphocytes were found within ectopic lymphoid tissue, activation-induced cytidine deaminase was expressed, and class-switched B cells were present. The presence of circular DNA intermediates, a hallmark of active class switch recombination, suggested that class switching occurs within the ectopic lymphoid tissue. Individual collections of ectopic lymphoid tissue ("lipogranulomas") from the same mouse contained different B cell repertoires, consistent with local germinal center-like reactions. Class-switched anti-RNP autoantibody-producing cells were also found in the lipogranulomas. Somatic hypermutation in the lipogranulomas was T cell-dependent, as was the production of isotype-switched anti-Sm/RNP autoantibodies. Thus, ectopic lymphoid tissue induced by TMPD recapitulates many of the functional characteristics of secondary lymphoid tissue and contains autoantibody-secreting cells, which may escape from normal censoring mechanisms in this location.


Subject(s)
Adipose Tissue/immunology , Adipose Tissue/pathology , B-Lymphocytes/immunology , Choristoma/immunology , Lupus Erythematosus, Systemic/immunology , Lymphoid Tissue , Animals , Autoantibodies/biosynthesis , Autoantigens/immunology , Cell Proliferation , Choristoma/pathology , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Germinal Center/immunology , Germinal Center/pathology , Immunoglobulin Class Switching/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , Immunohistochemistry , Immunosuppressive Agents/toxicity , Lupus Erythematosus, Systemic/chemically induced , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Inbred BALB C , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleoproteins, Small Nuclear/immunology , Somatic Hypermutation, Immunoglobulin/immunology , T-Lymphocytes/immunology , Terpenes/toxicity
17.
J Immunol ; 181(5): 3259-67, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18713997

ABSTRACT

Chronic inflammation promotes the formation of ectopic lymphoid tissue morphologically resembling secondary lymphoid tissues, though it is unclear whether this is a location where Ag-specific immune responses develop or merely a site of lymphocyte accumulation. Ectopic lymphoid tissue formation is associated with many humoral autoimmune diseases, including lupus induced by tetramethylpecadentane in mice. We examined whether an immune response to 4-hydroxy-3-nitrophenyl acetyl-keyhole limpet hemocyanin (NP-KLH) and NP-OVA develops within ectopic lymphoid tissue ("lipogranulomas") induced by tetramethylpecadentane in C57BL/6 mice. Following primary immunization, NP-specific B cells bearing V186.2 and related heavy chains as well as lambda-light chains accumulated within ectopic lymphoid tissue. The number of anti-NP-secreting B cells in the ectopic lymphoid tissue was greatly enhanced by immunization with NP-KLH. Remarkably, the H chain sequences isolated from individual lipogranulomas from these mice were diverse before immunization, whereas individual lipogranulomas from single immunized mice had unique oligo- or monoclonal populations of presumptive NP-specific B cells. H chain CDR sequences bore numerous replacement mutations, consistent with an Ag-driven and T cell-mediated response. In mice adoptively transferred with OT-II or DO11 T cells, there was a striking accumulation of OVA-specific T cells in lipogranulomas after s.c. immunization with NP-OVA. The selective colocalization of proliferating, Ag-specific T and B lymphocytes in lipogranulomas from tetramethylpecadentane-treated mice undergoing primary immunization implicates ectopic lymphoid tissue as a site where Ag-specific humoral immune responses can develop. This has implications for understanding the strong association of humoral autoimmunity with lymphoid neogenesis, which may be associated with deficient censoring of autoreactive cells.


Subject(s)
Antigens/immunology , B-Lymphocytes/immunology , Choristoma/pathology , Lymphoid Tissue/pathology , T-Lymphocytes/immunology , Animals , Antibody Formation , Autoimmunity , Female , Immunization , Immunoglobulin Heavy Chains , Immunoglobulin Light Chains , Mice
18.
iScience ; 23(5): 101050, 2020 May 22.
Article in English | MEDLINE | ID: mdl-32339990

ABSTRACT

Myeloid derived suppressor cells (MDSCs) are a population of immature myeloid cells that suppress adaptive immune function, yet the factors that regulate their suppressive function in patients with infection remain unclear. We studied MDSCs in patients with leprosy, a disease caused by Mycobacterium leprae, where clinical manifestations present on a spectrum that correlate with immunity to the pathogen. We found that HLA-DR-CD33+CD15+ MDSCs were increased in blood from patients with disseminated/progressive lepromatous leprosy and possessed T cell-suppressive activity as compared with self-limiting tuberculoid leprosy. Mechanistically, we found ER stress played a critical role in regulating the T cell suppressive activity in these MDSCs. Furthermore, ER stress augmented IL-10 production, contributing to MDSC activity, whereas IFN-γ allowed T cells to overcome MDSC suppressive activity. These studies highlight a regulatory mechanism that links ER stress to IL-10 in mediating MDSC suppressive function in human infectious disease.

19.
J Invest Dermatol ; 135(10): 2410-2417, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26030183

ABSTRACT

The mechanisms by which intracellular pathogens trigger immunosuppressive pathways are critical for understanding the pathogenesis of microbial infection. One pathway that inhibits host defense responses involves the induction of type I interferons and subsequently IL-10, yet the mechanism by which type I IFN induces IL-10 remains unclear. Our studies of gene expression profiles derived from leprosy skin lesions suggested a link between IL-27 and the IFN-ß induced IL-10 pathway. Here, we demonstrate that the IL-27p28 subunit is upregulated following treatment of monocytes with IFN-ß and Mycobacterium leprae, the intracellular bacterium that causes leprosy. The ability of IFN-ß and M. leprae to induce IL-10 was diminished by IL-27 knockdown. Additionally, treatment of monocytes with recombinant IL-27 was sufficient to induce the production of IL-10. Functionally, IL-27 inhibited the ability of IFN-γ to trigger antimicrobial activity against M. leprae in infected monocytes. At the site of disease, IL-27 was more strongly expressed in skin lesions of patients with progressive lepromatous leprosy, correlating and colocalizing with IFN-ß and IL-10 in macrophages. Together, these data provide evidence that in the human cutaneous immune responses to microbial infection, IL-27 contributes to the suppression of host antimicrobial responses.


Subject(s)
Interferon-beta/pharmacology , Interleukin-10/metabolism , Interleukin-27/metabolism , Leprosy, Lepromatous/drug therapy , Leprosy, Lepromatous/metabolism , Mycobacterium leprae/metabolism , Animals , Biomarkers/metabolism , Cells, Cultured , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Immunosuppressive Agents/pharmacology , Interleukin-27/pharmacology , Leprosy, Lepromatous/pathology , Mice , Microscopy, Confocal , Models, Animal , Monocytes/cytology , Monocytes/drug effects , Mycobacterium leprae/pathogenicity , Prognosis , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction/methods , Sampling Studies , Sensitivity and Specificity , Transfection
20.
J Exp Med ; 208(8): 1673-82, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21746813

ABSTRACT

Microbes activate pattern recognition receptors to initiate adaptive immunity. T cells affect early innate inflammatory responses to viral infection, but both activation and suppression have been demonstrated. We identify a novel role for B cells in the early innate immune response during bacterial sepsis. We demonstrate that Rag1(-/-) mice display deficient early inflammatory responses and reduced survival during sepsis. Interestingly, B cell-deficient or anti-CD20 B cell-depleted mice, but not α/ß T cell-deficient mice, display decreased inflammatory cytokine and chemokine production and reduced survival after sepsis. Both treatment of B cell-deficient mice with serum from wild-type (WT) mice and repletion of Rag1(-/-) mice with B cells improves sepsis survival, suggesting antibody-independent and antibody-dependent roles for B cells in the outcome to sepsis. During sepsis, marginal zone and follicular B cells are activated through type I interferon (IFN-I) receptor (IFN-α/ß receptor [IFNAR]), and repleting Rag1(-/-) mice with WT, but not IFNAR(-/-), B cells improves IFN-I-dependent and -independent early cytokine responses. Repleting B cell-deficient mice with the IFN-I-dependent chemokine, CXCL10 was also sufficient to improve sepsis survival. This study identifies a novel role for IFN-I-activated B cells in protective early innate immune responses during bacterial sepsis.


Subject(s)
B-Lymphocytes/immunology , Immunity, Innate/immunology , Sepsis/immunology , Analysis of Variance , Animals , B-Lymphocytes/metabolism , Chemokine CXCL10/immunology , Colony Count, Microbial , Flow Cytometry , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Mice , Mice, Knockout , Receptor, Interferon alpha-beta/immunology , Receptors, Pattern Recognition/immunology
SELECTION OF CITATIONS
SEARCH DETAIL