Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
JTCVS Open ; 18: 324-344, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38690424

ABSTRACT

Objective: Malignant pleural mesothelioma is a fatal disease and a clinical challenge, as few effective treatment modalities are available. Previous evidence links the gut microbiome to the host immunoreactivity to tumors. We thus evaluated the impact of a novel microbiome modulator compound (MMC) on the gut microbiota composition, tumor immune microenvironment, and cancer control in a model of malignant pleural mesothelioma. Methods: Age- and weight-matched immunocompetent (n = 23) or athymic BALB/c mice (n = 15) were randomly assigned to MMC or no treatment (control) groups. MMC (31 ppm) was administered through the drinking water 14 days before AB12 malignant mesothelioma cell inoculation into the pleural cavity. The impact of MMC on tumor growth, animal survival, tumor-infiltrating leucocytes, gut microbiome, and fecal metabolome was evaluated and compared with those of control animals. Results: The MMC delayed tumor growth and significantly prolonged the survival of immunocompetent animals (P = .0015) but not that of athymic mice. The improved tumor control in immunocompetent mice correlated with increased infiltration of CD3+CD8+GRZB+ cytotoxic T lymphocytes in tumors. Gut microbiota analyses indicated an enrichment in producers of short chain fatty acids in MMC-treated animals. Finally, we observed a positive correlation between the level of fecal short chain fatty acids and abundance of tumor-infiltrating cytotoxic T cells in malignant pleural mesothelioma. Conclusions: MMC administration boosts antitumor immunity, which correlates with a change in gut microbiome and metabolome. MMC may represent a valuable treatment option to combine with immunotherapy in patients with cancer.

2.
Mucosal Immunol ; 16(2): 194-207, 2023 04.
Article in English | MEDLINE | ID: mdl-36868478

ABSTRACT

Our skin has a unique barrier function, which is imperative for the body's protection against external pathogens and environmental insults. Although interacting closely and sharing many similarities with key mucosal barrier sites, such as the gut and the lung, the skin also provides protection for internal tissues and organs and has a distinct lipid and chemical composition. Skin immunity develops over time and is influenced by a multiplicity of different factors, including lifestyle, genetics, and environmental exposures. Alterations in early life skin immune and structural development may have long-term consequences for skin health. In this review, we summarize the current knowledge on cutaneous barrier and immune development from early life to adulthood, with an overview of skin physiology and immune responses. We specifically highlight the influence of the skin microenvironment and other host intrinsic, host extrinsic (e.g. skin microbiome), and environmental factors on early life cutaneous immunity.


Subject(s)
Microbiota , Skin , Mucous Membrane , Lung , Environmental Exposure/adverse effects
3.
Mucosal Immunol ; 15(5): 908-926, 2022 05.
Article in English | MEDLINE | ID: mdl-35672452

ABSTRACT

Barrier integrity is central to the maintenance of healthy immunological homeostasis. Impaired skin barrier function is linked with enhanced allergen sensitization and the development of diseases such as atopic dermatitis (AD), which can precede the development of other allergic disorders, for example, food allergies and asthma. Epidemiological evidence indicates that children suffering from allergies have lower levels of dietary fibre-derived short-chain fatty acids (SCFA). Using an experimental model of AD-like skin inflammation, we report that a fermentable fibre-rich diet alleviates systemic allergen sensitization and disease severity. The gut-skin axis underpins this phenomenon through SCFA production, particularly butyrate, which strengthens skin barrier function by altering mitochondrial metabolism of epidermal keratinocytes and the production of key structural components. Our results demonstrate that dietary fibre and SCFA improve epidermal barrier integrity, ultimately limiting early allergen sensitization and disease development.The Graphical Abstract was designed using Servier Medical Art images ( https://smart.servier.com ).


Subject(s)
Dermatitis, Atopic , Food Hypersensitivity , Allergens , Child , Dietary Fiber , Fatty Acids, Volatile , Humans , Keratinocytes
4.
Nat Commun ; 12(1): 2911, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34006859

ABSTRACT

The impact of immune mediators on weight homeostasis remains underdefined. Interrogation of resistance to diet-induced obesity in mice lacking a negative regulator of Toll-like receptor signaling serendipitously uncovered a role for B cell activating factor (BAFF). Here we show that overexpression of BAFF in multiple mouse models associates with protection from weight gain, approximating a log-linear dose response relation to BAFF concentrations. Gene expression analysis of BAFF-stimulated subcutaneous white adipocytes unveils upregulation of lipid metabolism pathways, with BAFF inducing white adipose tissue (WAT) lipolysis. Brown adipose tissue (BAT) from BAFF-overexpressing mice exhibits increased Ucp1 expression and BAFF promotes brown adipocyte respiration and in vivo energy expenditure. A proliferation-inducing ligand (APRIL), a BAFF homolog, similarly modulates WAT and BAT lipid handling. Genetic deletion of both BAFF and APRIL augments diet-induced obesity. Lastly, BAFF/APRIL effects are conserved in human adipocytes and higher BAFF/APRIL levels correlate with greater BMI decrease after bariatric surgery. Together, the BAFF/APRIL axis is a multifaceted immune regulator of weight gain and adipose tissue function.


Subject(s)
B-Cell Activating Factor/genetics , Obesity/genetics , Signal Transduction/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Weight Gain/genetics , Adipocytes/cytology , Adipocytes/metabolism , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Animals , B-Cell Activating Factor/metabolism , Cells, Cultured , Diet, High-Fat/adverse effects , Gene Expression Profiling/methods , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Obesity/etiology , Obesity/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism
5.
Nat Immunol ; 22(3): 279-286, 2021 03.
Article in English | MEDLINE | ID: mdl-33495652

ABSTRACT

The constituents of the gut microbiome are determined by the local habitat, which itself is shaped by immunological pressures, such as mucosal IgA. Using a mouse model of restricted antibody repertoire, we identified a role for antibody-microbe interactions in shaping a community of bacteria with an enhanced capacity to metabolize L-tyrosine. This model led to increased concentrations of p-cresol sulfate (PCS), which protected the host against allergic airway inflammation. PCS selectively reduced CCL20 production by airway epithelial cells due to an uncoupling of epidermal growth factor receptor (EGFR) and Toll-like receptor 4 (TLR4) signaling. Together, these data reveal a gut microbe-derived metabolite pathway that acts distally on the airway epithelium to reduce allergic airway responses, such as those underpinning asthma.


Subject(s)
Antibodies/metabolism , Bacteria/metabolism , Cresols/metabolism , Gastrointestinal Microbiome , Intestines/microbiology , Lung/metabolism , Pneumonia/prevention & control , Respiratory Hypersensitivity/prevention & control , Sulfuric Acid Esters/metabolism , Tyrosine/metabolism , Administration, Oral , Allergens , Animals , Antibodies/immunology , Antibody Diversity , Bacteria/immunology , Cells, Cultured , Chemokine CCL20/metabolism , Coculture Techniques , Cresols/administration & dosage , Disease Models, Animal , ErbB Receptors/metabolism , Female , Host-Pathogen Interactions , Injections, Intravenous , Lung/immunology , Lung/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/microbiology , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/metabolism , Respiratory Hypersensitivity/microbiology , Signal Transduction , Sulfuric Acid Esters/administration & dosage , Toll-Like Receptor 4/metabolism , Tyrosine/administration & dosage
6.
J Allergy Clin Immunol ; 147(3): 1049-1062.e7, 2021 03.
Article in English | MEDLINE | ID: mdl-32679208

ABSTRACT

BACKGROUND: Allergic skin inflammation often presents in early childhood; however, little is known about the events leading to its initiation and whether it is transient or long-term in nature. OBJECTIVE: We sought to determine the immunologic rules that govern skin inflammation in early life. METHODS: Neonatal and adult mice were epicutaneously sensitized with allergen followed by airway allergen challenge. Epicutaneous application of labeled allergen allowed for determination of antigen uptake and processing by antigen-presenting cells. RNAseq and microbiome analysis was performed on skin from neonatal and adult specific pathogen-free and germ-free mice. RESULTS: A mixed TH2/TH17 inflammatory response in the skin and the lungs of adult mice was observed following sensitization and challenge. Comparatively, neonatal mice did not develop overt skin inflammation, but exhibited systemic release of IL-17a and a TH2-dominated lung response. Mechanical skin barrier disruption was not sufficient to drive allergic skin inflammation, although it did promote systemic immune priming. Skin of neonatal mice and adult germ-free mice was seeded with low numbers of antigen-presenting cells and impaired chemokine and alarmin production. Enhanced chemokine and alarmin production, and seeding of the skin with antigen-presenting cells capable of instructing recruited cells to elicit their effector function, was, at least in part, dependent on formation of the microbiome, and consequently contributed to the development of overt skin disease. CONCLUSIONS: These data shed light on the principles that underlie allergic inflammation in different tissues and highlight a window of opportunity that might exist for early-life prevention of allergic diseases.


Subject(s)
Antigen-Presenting Cells/immunology , Hypersensitivity/immunology , Inflammation/immunology , Lung/immunology , Microbiota/immunology , Skin/immunology , Th2 Cells/immunology , Animals , Antigens, Dermatophagoides/immunology , Cell Movement , Chemokines/metabolism , Disease Models, Animal , Female , Germ-Free Life , Humans , Hypersensitivity/microbiology , Inflammation/microbiology , Interleukin-17/metabolism , Male , Mice , Mice, Inbred BALB C , Pyroglyphidae
7.
Sci Transl Med ; 12(540)2020 04 22.
Article in English | MEDLINE | ID: mdl-32321863

ABSTRACT

Eicosanoids are key mediators of type-2 inflammation, e.g., in allergy and asthma. Helminth products have been suggested as remedies against inflammatory diseases, but their effects on eicosanoids are unknown. Here, we show that larval products of the helminth Heligmosomoides polygyrus bakeri (HpbE), known to modulate type-2 responses, trigger a broad anti-inflammatory eicosanoid shift by suppressing the 5-lipoxygenase pathway, but inducing the cyclooxygenase (COX) pathway. In human macrophages and granulocytes, the HpbE-driven induction of the COX pathway resulted in the production of anti-inflammatory mediators [e.g., prostaglandin E2 (PGE2) and IL-10] and suppressed chemotaxis. HpbE also abrogated the chemotaxis of granulocytes from patients suffering from aspirin-exacerbated respiratory disease (AERD), a severe type-2 inflammatory condition. Intranasal treatment with HpbE extract attenuated allergic airway inflammation in mice, and intranasal transfer of HpbE-conditioned macrophages led to reduced airway eosinophilia in a COX/PGE2-dependent fashion. The induction of regulatory mediators in macrophages depended on p38 mitogen-activated protein kinase (MAPK), hypoxia-inducible factor-1α (HIF-1α), and Hpb glutamate dehydrogenase (GDH), which we identify as a major immunoregulatory protein in HpbE Hpb GDH activity was required for anti-inflammatory effects of HpbE in macrophages, and local administration of recombinant Hpb GDH to the airways abrogated allergic airway inflammation in mice. Thus, a metabolic enzyme present in helminth larvae can suppress type-2 inflammation by inducing an anti-inflammatory eicosanoid switch, which has important implications for the therapy of allergy and asthma.


Subject(s)
Eicosanoids , Helminths , Animals , Anti-Inflammatory Agents , Cyclooxygenase 2 , Humans , Inflammation , Larva , Mice
8.
Immunity ; 48(5): 992-1005.e8, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29768180

ABSTRACT

Dietary fiber protects against chronic inflammatory diseases by dampening immune responses through short-chain fatty acids (SCFAs). Here we examined the effect of dietary fiber in viral infection, where the anti-inflammatory properties of SCFAs in principle could prevent protective immunity. Instead, we found that fermentable dietary fiber increased survival of influenza-infected mice through two complementary mechanisms. High-fiber diet (HFD)-fed mice exhibited altered bone marrow hematopoiesis, characterized by enhanced generation of Ly6c- patrolling monocytes, which led to increased numbers of alternatively activated macrophages with a limited capacity to produce the chemokine CXCL1 in the airways. Blunted CXCL1 production reduced neutrophil recruitment to the airways, thus limiting tissue immunopathology during infection. In parallel, diet-derived SCFAs boosted CD8+ T cell effector function by enhancing cellular metabolism. Hence, dietary fermentable fiber and SCFAs set an immune equilibrium, balancing innate and adaptive immunity so as to promote the resolution of influenza infection while preventing immune-associated pathology.


Subject(s)
Antigens, Ly/immunology , CD8-Positive T-Lymphocytes/immunology , Dietary Fiber/pharmacology , Hematopoiesis/immunology , Monocytes/immunology , Orthomyxoviridae Infections/immunology , Adaptive Immunity/drug effects , Adaptive Immunity/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Dietary Fiber/administration & dosage , Fatty Acids, Volatile/immunology , Fatty Acids, Volatile/metabolism , Hematopoiesis/drug effects , Humans , Immunity, Innate/drug effects , Immunity, Innate/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects , Monocytes/metabolism , Protective Agents/administration & dosage , Protective Agents/pharmacology
9.
Am J Respir Crit Care Med ; 193(9): 975-87, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26630356

ABSTRACT

RATIONALE: Changes in the pulmonary microbiota are associated with progressive respiratory diseases including chronic obstructive pulmonary disease (COPD). Whether there is a causal relationship between these changes and disease progression remains unknown. OBJECTIVES: To investigate the link between an altered microbiota and disease, we used a murine model of chronic lung inflammation that is characterized by key pathological features found in COPD and compared responses in specific pathogen-free (SPF) mice and mice depleted of microbiota by antibiotic treatment or devoid of a microbiota (axenic). METHODS: Mice were challenged with LPS/elastase intranasally over 4 weeks, resulting in a chronically inflamed and damaged lung. The ensuing cellular infiltration, histological damage, and decline in lung function were quantified. MEASUREMENTS AND MAIN RESULTS: Similar to human disease, the composition of the pulmonary microbiota was altered in diseased animals. We found that the microbiota richness and diversity were decreased in LPS/elastase-treated mice, with an increased representation of the genera Pseudomonas and Lactobacillus and a reduction in Prevotella. Moreover, the microbiota was implicated in disease development as mice depleted, or devoid, of microbiota exhibited an improvement in lung function, reduced inflammation, and lymphoid neogenesis. The absence of microbial cues markedly decreased the production of IL-17A, whereas intranasal transfer of fluid enriched with the pulmonary microbiota isolated from diseased mice enhanced IL-17A production in the lungs of antibiotic-treated or axenic recipients. Finally, in mice harboring a microbiota, neutralizing IL-17A dampened inflammation and restored lung function. CONCLUSIONS: Collectively, our data indicate that host-microbial cross-talk promotes inflammation and could underlie the chronicity of inflammatory lung diseases.


Subject(s)
Autoantibodies/immunology , Inflammation/physiopathology , Interleukin-17/immunology , Microbiota , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/physiopathology , Animals , Disease Models, Animal , Inflammation/complications , Inflammation/immunology , Lung/immunology , Lung/physiopathology , Mice , Mice, Inbred BALB C , Pulmonary Disease, Chronic Obstructive/complications
10.
Ann Am Thorac Soc ; 12 Suppl 2: S150-6, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26595731

ABSTRACT

Host-microorganism interactions shape local cell functionality, immune responses, and can influence disease development. Evidence indicates that the impact of host-microbe interactions reaches far beyond the local environment, thus influencing responses in peripheral tissues. There is a vital cross-talk between the mucosal tissues of our body, as exemplified by intestinal complications during respiratory disease and vice versa. Although, mechanistically, this phenomenon remains poorly defined, the existence of the gut-lung axis and its implications in both health and disease could be profoundly important for both disease etiology and treatment. In this review, we highlight how changes in the intestinal microenvironment, with a particular focus on the intestinal microbiota, impact upon respiratory disease.


Subject(s)
Host-Pathogen Interactions , Intestinal Mucosa/microbiology , Microbiota , Respiratory Mucosa/microbiology , Respiratory Tract Diseases/microbiology , Humans , Probiotics/therapeutic use , Respiratory Tract Diseases/drug therapy
11.
PLoS One ; 9(2): e98440, 2014.
Article in English | MEDLINE | ID: mdl-24918427

ABSTRACT

For patients with chronic lung diseases, such as chronic obstructive pulmonary disease (COPD), exacerbations are life-threatening events causing acute respiratory distress that can even lead to hospitalization and death. Although a great deal of effort has been put into research of exacerbations and potential treatment options, the exact underlying mechanisms are yet to be deciphered and no therapy that effectively targets the excessive inflammation is available. In this study, we report that interleukin-1ß (IL-1ß) and interleukin-17A (IL-17A) are key mediators of neutrophilic inflammation in influenza-induced exacerbations of chronic lung inflammation. Using a mouse model of disease, our data shows a role for IL-1ß in mediating lung dysfunction, and in driving neutrophilic inflammation during the whole phase of viral infection. We further report a role for IL-17A as a mediator of IL-1ß induced neutrophilia at early time points during influenza-induced exacerbations. Blocking of IL-17A or IL-1 resulted in a significant abrogation of neutrophil recruitment to the airways in the initial phase of infection or at the peak of viral replication, respectively. Therefore, IL-17A and IL-1ß are potential targets for therapeutic treatment of viral exacerbations of chronic lung inflammation.


Subject(s)
Influenza, Human/complications , Interleukin-17/immunology , Interleukin-1beta/immunology , Neutrophil Infiltration , Orthomyxoviridae Infections/complications , Pneumonia/complications , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , Chronic Disease , Humans , Influenza, Human/immunology , Influenza, Human/therapy , Interleukin 1 Receptor Antagonist Protein/immunology , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Interleukin-17/antagonists & inhibitors , Interleukin-1beta/antagonists & inhibitors , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/therapy , Pneumonia/immunology , Pneumonia/therapy , Pulmonary Disease, Chronic Obstructive/complications , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/therapy , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use
12.
Nat Med ; 20(6): 642-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24813249

ABSTRACT

Epidemiological data point toward a critical period in early life during which environmental cues can set an individual on a trajectory toward respiratory health or disease. The neonatal immune system matures during this period, although little is known about the signals that lead to its maturation. Here we report that the formation of the lung microbiota is a key parameter in this process. Immediately following birth, neonatal mice were prone to develop exaggerated airway eosinophilia, release type 2 helper T cell cytokines and exhibit airway hyper-responsiveness following exposure to house dust mite allergens, even though their lungs harbored high numbers of natural CD4(+)Foxp3(+)CD25(+)Helios(+) regulatory T (Treg) cells. During the first 2 weeks after birth, the bacterial load in the lungs increased, and representation of the bacterial phyla shifts from a predominance of Gammaproteobacteria and Firmicutes towards Bacteroidetes. The changes in the microbiota were associated with decreased aeroallergen responsiveness and the emergence of a Helios(-) Treg cell subset that required interaction with programmed death ligand 1 (PD-L1) for development. Absence of microbial colonization(10) or blockade of PD-L1 during the first 2 weeks postpartum maintained exaggerated responsiveness to allergens through to adulthood. Adoptive transfer of Treg cells from adult mice to neonates before aeroallergen exposure ameliorated disease. Thus, formation of the airway microbiota induces regulatory cells early in life, which, when dysregulated, can lead to sustained susceptibility to allergic airway inflammation in adulthood.


Subject(s)
Allergens/immunology , Animals, Newborn/microbiology , B7-H1 Antigen/immunology , Immune Tolerance/immunology , Lung/immunology , Lung/microbiology , Microbiota/immunology , Adoptive Transfer , Animals , Animals, Newborn/immunology , Bronchial Provocation Tests , Bronchoalveolar Lavage Fluid/chemistry , Cytokines/metabolism , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Mice , Mice, Inbred BALB C , Pyroglyphidae/immunology , Real-Time Polymerase Chain Reaction , T-Lymphocytes, Regulatory/immunology
13.
Nat Med ; 20(2): 159-66, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24390308

ABSTRACT

Metabolites from intestinal microbiota are key determinants of host-microbe mutualism and, consequently, the health or disease of the intestinal tract. However, whether such host-microbe crosstalk influences inflammation in peripheral tissues, such as the lung, is poorly understood. We found that dietary fermentable fiber content changed the composition of the gut and lung microbiota, in particular by altering the ratio of Firmicutes to Bacteroidetes. The gut microbiota metabolized the fiber, consequently increasing the concentration of circulating short-chain fatty acids (SCFAs). Mice fed a high-fiber diet had increased circulating levels of SCFAs and were protected against allergic inflammation in the lung, whereas a low-fiber diet decreased levels of SCFAs and increased allergic airway disease. Treatment of mice with the SCFA propionate led to alterations in bone marrow hematopoiesis that were characterized by enhanced generation of macrophage and dendritic cell (DC) precursors and subsequent seeding of the lungs by DCs with high phagocytic capacity but an impaired ability to promote T helper type 2 (TH2) cell effector function. The effects of propionate on allergic inflammation were dependent on G protein-coupled receptor 41 (GPR41, also called free fatty acid receptor 3 or FFAR3), but not GPR43 (also called free fatty acid receptor 2 or FFAR2). Our results show that dietary fermentable fiber and SCFAs can shape the immunological environment in the lung and influence the severity of allergic inflammation.


Subject(s)
Bacteroidetes/metabolism , Dietary Fiber/microbiology , Hematopoiesis/physiology , Hypersensitivity/physiopathology , Intestines/microbiology , Microbiota/physiology , Adoptive Transfer , Animals , Base Sequence , Cytokines/metabolism , DNA Primers/genetics , DNA, Bacterial/isolation & purification , Enzyme-Linked Immunosorbent Assay , Fatty Acids/blood , Feces/chemistry , Female , Flow Cytometry , Lung/chemistry , Lung/pathology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Statistics, Nonparametric
14.
Front Med (Lausanne) ; 1: 41, 2014.
Article in English | MEDLINE | ID: mdl-25593914

ABSTRACT

Secondary bacterial infections following influenza infection are a pressing problem facing respiratory medicine. Although antibiotic treatment has been highly successful over recent decades, fatalities due to secondary bacterial infections remain one of the leading causes of death associated with influenza. We have assessed whether administration of a bacterial extract alone is sufficient to potentiate immune responses and protect against primary infection with influenza, and secondary infections with either Streptococcus pneumoniae or Klebsiella pneumoniae in mice. We show that oral administration with the bacterial extract, OM-85, leads to a maturation of dendritic cells and B-cells characterized by increases in MHC II, CD86, and CD40, and a reduction in ICOSL. Improved immune responsiveness against influenza virus reduced the threshold of susceptibility to secondary bacterial infections, and thus protected the mice. The protection was associated with enhanced polyclonal B-cell activation and release of antibodies that were effective at neutralizing the virus. Taken together, these data show that oral administration of bacterial extracts provides sufficient mucosal immune stimulation to protect mice against a respiratory tract viral infection and associated sequelae.

15.
J Infect Dis ; 205(1): 152-61, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21990421

ABSTRACT

Recent studies have underscored physiological and pathophysiological roles for the tryptophan-degrading enzyme indolamine 2,3-dioxygenase (IDO) in immune counterregulation. However, IDO was first recognized as an antimicrobial effector, restricting tryptophan availability to Toxoplasma gondii and other pathogens in vitro. The biological relevance of these findings came under question when infectious phenotypes were not forthcoming in IDO-deficient mice. The recent discovery of an IDO homolog, IDO-2, suggested that the issue deserved reexamination. IDO inhibition during murine toxoplasmosis led to 100% mortality, with increased parasite burdens and no evident effects on the immune response. Similar studies revealed a counterregulatory role for IDO during leishmaniasis (restraining effector immune responses and parasite clearance), and no evident role for IDO in herpes simplex virus type 1 (HSV-1) infection. Thus, IDO plays biologically important roles in the host response to diverse intracellular infections, but the dominant nature of this role--antimicrobial or immunoregulatory--is pathogen-specific.


Subject(s)
Herpes Simplex/enzymology , Herpesvirus 1, Human , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Leishmaniasis, Cutaneous/immunology , Toxoplasmosis, Animal/immunology , Animals , Female , Herpes Simplex/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Leishmaniasis, Cutaneous/enzymology , Male , Mice , Mice, Inbred C57BL , Toxoplasmosis, Animal/enzymology , Tryptophan/analogs & derivatives , Tryptophan/metabolism
16.
PLoS Negl Trop Dis ; 5(9): e1316, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21909452

ABSTRACT

BACKGROUND: Leishmaniasis remains a significant cause of morbidity and mortality in the tropics. Available therapies are problematic due to toxicity, treatment duration and emerging drug resistance. Mouse models of leishmaniasis have demonstrated that disease outcome depends critically on the balance between effector and regulatory CD4(+) T cell responses, something mirrored in descriptive studies of human disease. Recombinant IL-2/diphtheria toxin fusion protein (rIL-2/DTx), a drug that is FDA-approved for the treatment of cutaneous T cell lymphoma, has been reported to deplete regulatory CD4(+) T cells. METHODOLOGY/PRINCIPAL FINDINGS: We investigated the potential efficacy of rIL-2/DTx as adjunctive therapy for experimental infection with Leishmania major. Treatment with rIL-2/DTx suppressed lesional regulatory T cell numbers and was associated with significantly increased antigen-specific IFN-γ production, enhanced lesion resolution and decreased parasite burden. Combined administration of rIL-2/DTx and sodium stibogluconate had additive biological and therapeutic effects, allowing for reduced duration or dose of sodium stibogluconate therapy. CONCLUSIONS/SIGNIFICANCE: These data suggest that pharmacological suppression of immune counterregulation using a commercially available drug originally developed for cancer therapy may have practical therapeutic utility in leishmaniasis. Rational reinvestigation of the efficacy of drugs approved for other indications in experimental models of neglected tropical diseases has promise in providing new candidates to the drug discovery pipeline.


Subject(s)
Antimony Sodium Gluconate/administration & dosage , Antiprotozoal Agents/administration & dosage , Biological Products/administration & dosage , Diphtheria Toxin/administration & dosage , Immunologic Factors/administration & dosage , Interleukin-2/administration & dosage , Leishmaniasis, Cutaneous/drug therapy , Animals , Drug Therapy, Combination/methods , Humans , Leishmaniasis, Cutaneous/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Treatment Outcome
17.
J Immunol ; 184(2): 877-85, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20018611

ABSTRACT

IFN-gamma has long been recognized as a cytokine with potent and varied effects in the immune response. Although its effects on specific cell types have been well studied in vitro, its in vivo effects are less clearly understood because of its diverse actions on many different cell types. Although control of multiple protozoan parasites is thought to depend critically on the direct action of IFN-gamma on macrophages, this premise has never been directly proven in vivo. To more directly examine the effects of IFN-gamma on cells of the macrophage lineage in vivo, we generated mice called the "macrophages insensitive to IFN-gamma" (MIIG) mice, which express a dominant negative mutant IFN-gamma receptor in CD68+ cells: monocytes, macrophages, dendritic cells, and mast cells. Macrophage lineage cells and mast cells from these mice are unable to respond to IFN-gamma, whereas other cells are able to produce and respond to this cytokine normally. When challenged in vitro, macrophages from MIIG mice were unable produce NO or kill Trypanosoma cruzi or Leishmania major after priming with IFN-gamma. Furthermore, MIIG mice demonstrated impaired parasite control and heightened mortality after T. cruzi, L. major, and Toxoplasma gondii infection, despite an appropriate IFN-gamma response. In contrast, MIIG mice displayed normal control of lymphocytic choriomeningitis virus, despite persistent insensitivity of macrophages to IFN-gamma. Thus, the MIIG mouse formally demonstrates for the first time in vivo, the specific importance of direct, IFN-gamma mediated activation of macrophages for controlling infection with multiple protozoan parasites.


Subject(s)
Interferon-gamma/metabolism , Macrophages/metabolism , Protozoan Infections/immunology , Animals , Cell Lineage , Leishmania major , Macrophages/parasitology , Mice , Mice, Mutant Strains , Receptors, Interferon/genetics , Signal Transduction , Toxoplasma , Trypanosoma cruzi , Interferon gamma Receptor
18.
J Immunol ; 183(4): 2312-20, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19620304

ABSTRACT

IL-10 plays a central role in restraining the vigor of inflammatory responses, but the critical cellular sources of this counter-regulatory cytokine remain speculative in many disease models. Using a novel IL-10 transcriptional reporter mouse, we found an unexpected predominance of B cells (including plasma cells) among IL-10-expressing cells in peripheral lymphoid tissues at baseline and during diverse models of in vivo immunological challenge. Use of a novel B cell-specific IL-10 knockout mouse revealed that B cell-derived IL-10 nonredundantly decreases virus-specific CD8(+) T cell responses and plasma cell expansion during murine cytomegalovirus infection and modestly restrains immune activation after challenge with foreign Abs to IgD. In contrast, no role for B cell-derived IL-10 was evident during endotoxemia; however, although B cells dominated lymphoid tissue IL-10 production in this model, myeloid cells were dominant in blood and liver. These data suggest that B cells are an underappreciated source of counter-regulatory IL-10 production in lymphoid tissues, provide a clear rationale for testing the biological role of B cell-derived IL-10 in infectious and inflammatory disease, and underscore the utility of cell type-specific knockouts for mechanistic limning of immune counter-regulation.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Interleukin-10/physiology , Animals , B-Lymphocyte Subsets/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Disease Models, Animal , Female , Herpesviridae Infections/immunology , Herpesviridae Infections/metabolism , Herpesviridae Infections/pathology , Inflammation Mediators/physiology , Interleukin-10/biosynthesis , Interleukin-10/deficiency , Interleukin-10/genetics , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Lymphoid Tissue/immunology , Lymphoid Tissue/pathology , Lymphoid Tissue/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Muromegalovirus/immunology , NIH 3T3 Cells
19.
Nature ; 457(7229): 585-8, 2009 Jan 29.
Article in English | MEDLINE | ID: mdl-19060881

ABSTRACT

Aeroallergy results from maladaptive immune responses to ubiquitous, otherwise innocuous environmental proteins. Although the proteins targeted by aeroallergic responses represent a tiny fraction of the airborne proteins humans are exposed to, allergenicity is a quite public phenomenon-the same proteins typically behave as aeroallergens across the human population. Why particular proteins tend to act as allergens in susceptible hosts is a fundamental mechanistic question that remains largely unanswered. The main house-dust-mite allergen, Der p 2, has structural homology with MD-2 (also known as LY96), the lipopolysaccharide (LPS)-binding component of the Toll-like receptor (TLR) 4 signalling complex. Here we show that Der p 2 also has functional homology, facilitating signalling through direct interactions with the TLR4 complex, and reconstituting LPS-driven TLR4 signalling in the absence of MD-2. Mirroring this, airway sensitization and challenge with Der p 2 led to experimental allergic asthma in wild type and MD-2-deficient, but not TLR4-deficient, mice. Our results indicate that Der p 2 tends to be targeted by adaptive immune responses because of its auto-adjuvant properties. The fact that other members of the MD-2-like lipid-binding family are allergens, and that most defined major allergens are thought to be lipid-binding proteins, suggests that intrinsic adjuvant activity by such proteins and their accompanying lipid cargo may have some generality as a mechanism underlying the phenomenon of allergenicity.


Subject(s)
Allergens/immunology , Allergens/metabolism , Antigens, Dermatophagoides/immunology , Antigens, Dermatophagoides/metabolism , Molecular Mimicry/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Air , Allergens/chemistry , Allergens/genetics , Animals , Antigens, Dermatophagoides/chemistry , Antigens, Dermatophagoides/genetics , Arthropod Proteins , Asthma/genetics , Asthma/immunology , Cell Line , Disease Models, Animal , Female , Humans , Lipopolysaccharides/immunology , Lymphocyte Antigen 96/chemistry , Lymphocyte Antigen 96/deficiency , Lymphocyte Antigen 96/genetics , Lymphocyte Antigen 96/immunology , Lymphocyte Antigen 96/metabolism , Mice , Protein Binding , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
20.
J Leukoc Biol ; 82(2): 265-71, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17470533

ABSTRACT

As all immune responses have potential for damaging the host, tight regulation of such responses--in amplitude, space, time and character--is essential for maintaining health and homeostasis. It was thus inevitable that the initial wave of papers on the role of Toll-like receptors (TLRs), NOD-like receptors (NLRs) and RIG-I-like receptors (RLRs) in activating innate and adaptive immune responses would be followed by a second wave of reports focusing on the mechanisms responsible for restraining and modulating signaling by these receptors. This overview outlines current knowledge and controversies about the immunobiology of the RP105/MD-1 complex, a modulator of the most robustly signaling TLR, TLR4.


Subject(s)
Antigens, CD/physiology , Signal Transduction , Toll-Like Receptor 4/physiology , Animals , Antigens, CD/immunology , Gene Expression Regulation , Humans , Toll-Like Receptor 4/genetics
SELECTION OF CITATIONS
SEARCH DETAIL