Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Front Cell Infect Microbiol ; 12: 811474, 2022.
Article in English | MEDLINE | ID: mdl-35548467

ABSTRACT

Cryptococcosis is an invasive mycosis caused by Cryptococcus spp. that affects the lungs and the central nervous system (CNS). Due to the severity of the disease, it may occur concomitantly with other pathogens, as a coinfection. Pseudomonas aeruginosa (Pa), an opportunistic pathogen, can also cause pneumonia. In this work, we studied the interaction of C. gattii (Cg) and Pa, both in vitro and in vivo. Pa reduced growth of Cg by the secretion of inhibitory molecules in vitro. Macrophages previously stimulated with Pa presented increased fungicidal activity. In vivo, previous Pa infection reduced morbidity and delayed the lethality due to cryptococcosis. This phenotype was correlated with the decreased fungal burden in the lungs and brain, showing a delay of Cg translocation to the CNS. Also, there was increased production of IL-1ß, CXCL-1, and IL-10, together with the influx of iNOS-positive macrophages and neutrophils to the lungs. Altogether, Pa turned the lung into a hostile environment to the growth of a secondary pathogen, making it difficult for the fungus to translocate to the CNS. Further, iNOS inhibition reverted the Pa protective phenotype, suggesting its important role in the coinfection. Altogether, the primary Pa infection leads to balanced pro-inflammatory and anti-inflammatory responses during Cg infection. This response provided better control of cryptococcosis and was decisive for the mild evolution of the disease and prolonged survival of coinfected mice in a mechanism dependent on iNOS.


Subject(s)
Coinfection , Cryptococcosis , Cryptococcus gattii , Cryptococcus neoformans , Pseudomonas Infections , Animals , Cryptococcosis/microbiology , Mice , Phagocytosis
2.
Immunology ; 165(3): 355-368, 2022 03.
Article in English | MEDLINE | ID: mdl-34964126

ABSTRACT

Mucositis is a major clinical complication associated with cancer treatment and may limit the benefit of chemotherapy. Leukocytes and inflammatory mediators have been extensively associated with mucositis severity. However, the role of eosinophils in the pathophysiology of chemotherapy-induced mucositis remains to be elucidated. Here, using GATA-1-deficient mice, we investigated the role of eosinophils in intestinal mucositis. There was marked accumulation of eosinophils in mice given irinotecan and eosinophil ablation inhibited intestinal mucositis. Treatment with Evasin-4, a chemokine receptor antagonist, reduced the recruitment of eosinophils and decreased irinotecan-induced mucositis. Importantly, Evasin-4 did not interfere negatively with the antitumour effects of irinotecan. Evasin-4 was of benefit for mice given high doses of irinotecan once Evasin-4-treated mice presented delayed mortality. Altogether, our findings suggest that Evasin-4 may have significant mucosal-protective effects in the context of antineoplastic chemotherapy and may, therefore, be useful in combination with anticancer treatment in cancer patients.


Subject(s)
Antineoplastic Agents , Mucositis , Animals , Antineoplastic Agents/therapeutic use , Camptothecin/adverse effects , Eosinophils/pathology , Humans , Intestinal Mucosa/pathology , Irinotecan/adverse effects , Mice , Mucositis/chemically induced , Mucositis/drug therapy , Mucositis/pathology
3.
Gut Microbes ; 11(6): 1531-1546, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32573321

ABSTRACT

Although dysbiosis in the gut microbiota is known to be involved in several inflammatory diseases, whether any specific bacterial taxa control host response to inflammatory stimuli is still elusive. Here, we hypothesized that dysbiotic indigenous taxa could be involved in modulating host response to inflammatory triggers. To test this hypothesis, we conducted experiments in germ-free (GF) mice and in mice colonized with dysbiotic taxa identified in conventional (CV) mice subjected to chemotherapy-induced mucositis. First, we report that the absence of microbiota decreased inflammation and damage in the small intestine after administration of the chemotherapeutic agent 5-fluorouracil (5-FU). Also, 5-FU induced a shift in CV microbiota resulting in higher amounts of Enterobacteriaceae, including E. coli, in feces and small intestine and tissue damage. Prevention of Enterobacteriaceae outgrowth by treating mice with ciprofloxacin resulted in diminished 5-FU-induced tissue damage, indicating that this bacterial group is necessary for 5-FU-induced inflammatory response. In addition, monocolonization of germ-free (GF) mice with E. coli led to reversal of the protective phenotype during 5-FU chemotherapy. E. coli monocolonization decreased the basal plasma corticosterone levels and blockade of glucocorticoid receptor in GF mice restored inflammation upon 5-FU treatment. In contrast, treatment of CV mice with ciprofloxacin, that presented reduction of Enterobacteriaceae and E. coli content, induced an increase in corticosterone levels. Altogether, these findings demonstrate that Enterobacteriaceae outgrowth during dysbiosis impacts inflammation and tissue injury in the small intestine. Importantly, indigenous Enterobacteriaceae modulates host production of the anti-inflammatory steroid corticosterone and, consequently, controls inflammatory responsiveness in mice.


Subject(s)
Corticosterone/metabolism , Dysbiosis/microbiology , Enterobacteriaceae/growth & development , Animals , Antineoplastic Agents/adverse effects , Bacteria/classification , Bacteria/genetics , Bacteria/growth & development , Bacteria/isolation & purification , Corticosterone/immunology , Dysbiosis/etiology , Dysbiosis/immunology , Dysbiosis/metabolism , Enterobacteriaceae/genetics , Fluorouracil/adverse effects , Gastrointestinal Microbiome/drug effects , Humans , Intestine, Small/immunology , Intestine, Small/metabolism , Intestine, Small/microbiology , Male , Mice
4.
Cell Microbiol ; 22(6): e13179, 2020 06.
Article in English | MEDLINE | ID: mdl-32017324

ABSTRACT

Cryptococcus gattii (Cg) is one of the agents of cryptococcosis, a severe systemic mycosis with a higher prevalence in men than women, but the influence of the female sex hormone, 17-ß-estradiol (E2), on cryptococcosis remains unclear. Our study shows that female mice presented delayed mortality, increased neutrophil recruitment in bronchoalveolar lavage fluid, and reduced fungal load after 24 hr of infection compared to male and ovariectomised female mice (OVX). E2 replacement restored OVX female survival. Female macrophages have more efficient fungicidal activity, which was increased by E2 and reversed by the antagonist of G-protein-coupled oestrogen receptor (GPER), which negatively modulates PI3K activation. Furthermore, E2 induces a reduction in Cg cell diameter, cell charge, and antioxidant peroxidase activity. In conclusion, female mice present improved control of Cg infection, and GPER is important for E2 modulation of the female response.


Subject(s)
Cryptococcosis/drug therapy , Cryptococcus gattii/drug effects , Estradiol/pharmacology , GTP-Binding Proteins/metabolism , Macrophages/drug effects , Receptors, Estrogen/metabolism , Animals , Antifungal Agents/pharmacology , Antioxidants , Cryptococcosis/immunology , Disease Models, Animal , Female , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL
5.
Immunology ; 155(4): 477-490, 2018 12.
Article in English | MEDLINE | ID: mdl-30098206

ABSTRACT

The excessive inflammation often present in patients with severe dengue infection is considered both a hallmark of disease and a target for potential treatments. Interleukin-33 (IL-33) is a pleiotropic cytokine with pro-inflammatory effects whose role in dengue has not been fully elucidated. We demonstrate that IL-33 plays a disease-exacerbating role during experimental dengue infection in immunocompetent mice. Mice infected with dengue virus serotype 2 (DENV2) produced high levels of IL-33. DENV2-infected mice treated with recombinant IL-33 developed markedly more severe disease compared with untreated mice as assessed by mortality, granulocytosis, liver damage and pro-inflammatory cytokine production. Conversely, ST2-/- mice (deficient in IL-33 receptor) infected with DENV2 developed significantly less severe disease compared with wild-type mice. Furthermore, the increased disease severity and the accompanying pathology induced by IL-33 during dengue infection were reversed by the simultaneous treatment with a CXCR2 receptor antagonist (DF2156A). Together, these results indicate that IL-33 plays a disease-exacerbating role in experimental dengue infection, probably driven by CXCR2-expressing cells, leading to elevated pro-inflammatory response-mediated pathology. Our results also indicate that IL-33 is a potential therapeutic target for dengue infection.


Subject(s)
Dengue Virus/immunology , Interleukin-33/pharmacology , Receptors, Interleukin-8B/antagonists & inhibitors , Recombinant Proteins/pharmacology , Animals , Dengue/immunology , Dengue/virology , Disease Progression , Interleukin-1 Receptor-Like 1 Protein/deficiency , Interleukin-1 Receptor-Like 1 Protein/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sulfonamides/pharmacology
6.
Article in English | MEDLINE | ID: mdl-29018774

ABSTRACT

Influenza A virus (IAV) infects millions of people annually and predisposes to secondary bacterial infections. Inhalation of fungi within the Cryptococcus complex causes pulmonary disease with secondary meningo-encephalitis. Underlying pulmonary disease is a strong risk factor for development of C. gattii cryptococcosis though the effect of concurrent infection with IAV has not been studied. We developed an in vivo model of Influenza A H1N1 and C. gattii co-infection. Co-infection resulted in a major increase in morbidity and mortality, with severe lung damage and a high brain fungal burden when mice were infected in the acute phase of influenza multiplication. Furthermore, IAV alters the host response to C. gattii, leading to recruitment of significantly more neutrophils and macrophages into the lungs. Moreover, IAV induced the production of type 1 interferons (IFN-α4/ß) and the levels of IFN-γ were significantly reduced, which can be associated with impairment of the immune response to Cryptococcus during co-infection. Phagocytosis, killing of cryptococci and production of reactive oxygen species (ROS) by IAV-infected macrophages were reduced, independent of previous IFN-γ stimulation, leading to increased proliferation of the fungus within macrophages. In conclusion, IAV infection is a predisposing factor for severe disease and adverse outcomes in mice co-infected with C. gattii.


Subject(s)
Causality , Coinfection , Cryptococcosis/complications , Cryptococcus gattii/pathogenicity , Influenza A Virus, H1N1 Subtype/pathogenicity , Orthomyxoviridae Infections/complications , Acetylglucosaminidase/metabolism , Animals , Behavior, Animal , Brain/microbiology , Brain/pathology , Cell Proliferation , Chemokines/metabolism , Coinfection/immunology , Coinfection/microbiology , Coinfection/mortality , Coinfection/virology , Cryptococcosis/immunology , Cryptococcus gattii/immunology , Cryptococcus neoformans/immunology , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Dogs , Female , Humans , Influenza A Virus, H1N1 Subtype/immunology , Interferon-gamma/metabolism , Lung/enzymology , Lung/pathology , Lung/virology , Macrophages/metabolism , Macrophages/virology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL , Neutrophils , Nitric Oxide/metabolism , Orthomyxoviridae Infections/immunology , Peroxidase/metabolism , Peroxynitrous Acid/metabolism , Phagocytosis , Reactive Oxygen Species/metabolism , Survival Rate
7.
J Immunol ; 198(10): 4096-4106, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28424241

ABSTRACT

The indigenous intestinal microbiota is frequently considered an additional major organ of the human body and exerts profound immunomodulating activities. Germ-free (GF) mice display a significantly different inflammatory responsiveness pattern compared with conventional (CV) mice, and this was dubbed a "hyporesponsive phenotype." Taking into account that the deposition of immune complexes is a major event in acute inflammation and that GF mice have a distinct Ig repertoire and B cell activity, we aimed to evaluate whether this altered Ig repertoire interferes with the inflammatory responsiveness of GF mice. We found that serum transfer from CV naive mice was capable of reversing the inflammatory hyporesponsiveness of GF mice in sterile inflammatory injury induced by intestinal ischemia and reperfusion, as well as in a model of lung infection by Klebsiella pneumoniae Transferring serum from Ig-deficient mice to GF animals did not alter their response to inflammatory insult; however, injecting purified Abs from CV animals restored inflammatory responsiveness in GF mice, suggesting that natural Abs present in serum were responsible for altering GF responsiveness. Mechanistically, injection of serum and Ig from CV mice into GF animals restored IgG deposition, leukocyte influx, NF-κB activation, and proinflammatory gene expression in inflamed tissues and concomitantly downregulated annexin-1 and IL-10 production. Thus, our data show that microbiota-induced natural Abs are pivotal for host inflammatory responsiveness to sterile and infectious insults.


Subject(s)
Antibodies/immunology , Gastrointestinal Microbiome/immunology , Germ-Free Life , Inflammation/immunology , Intestines/immunology , Animals , Annexins/immunology , Antibodies/administration & dosage , B-Lymphocytes/immunology , Gene Expression Regulation , Humans , Interleukin-10/immunology , Intestines/microbiology , Intestines/pathology , Ischemia , Klebsiella Infections/immunology , Klebsiella Infections/microbiology , Klebsiella pneumoniae/immunology , Lung/immunology , Lung/microbiology , Mice , NF-kappa B/genetics
8.
Methods Mol Biol ; 1390: 301-16, 2016.
Article in English | MEDLINE | ID: mdl-26803637

ABSTRACT

The importance of the 24-h daily cycle, termed circadian, on immune function has been highlighted by a number of recent studies. Immune parameters such as the response to bacterial challenge or immune cell trafficking change with time of day and disruption of circadian rhythms has been linked to inflammatory pathologies. We are beginning to uncover that the key proteins that comprise the molecular clock, most notably BMAL1, CLOCK, and REV-ERBα, also control fundamental aspects of the immune response. Given the ubiquitous nature of the molecular clock in controlling many other types of physiologies such as metabolism and cardiovascular function, a more thorough understanding of the daily rhythm of the immune system may provide important insight into aspects of patient care such as vaccinations and how we manage infectious and inflammatory diseases. In this chapter, we describe a series of experiments to look at circadian expression and function in immune cells. The experiments described herein may provide an initial assessment of the role of the molecular clock on an immune response from any cell type of interest.


Subject(s)
Circadian Clocks/genetics , Circadian Clocks/immunology , Gene Expression Regulation , Immunity, Innate , Animals , CD11c Antigen/metabolism , Dexamethasone/pharmacology , Leukocytes/immunology , Leukocytes/metabolism , Macrophage Colony-Stimulating Factor/biosynthesis , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Reverse Transcriptase Polymerase Chain Reaction
9.
Immunology ; 146(3): 349-58, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26301993

ABSTRACT

Development of inflammatory diseases, such as metabolic syndrome and cancer, is prevalent in individuals that encounter continuous disruption of their internal clock. Further, daily oscillations in susceptibility to infection as well as a multitude of other immunological processes have been described. Much progress has been made and various mechanisms have been proposed to explain circadian variations in immunity; yet much is still unknown. Understanding the crosstalk between the circadian and the immune systems will allow us to manipulate clock outputs to prevent and treat inflammatory diseases in individuals at risk. This review briefly summarizes current knowledge about circadian rhythms and their role in the immune system and highlights progress and challenges in chrono-immunological research.


Subject(s)
Circadian Rhythm/immunology , Immune System/physiology , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/immunology , ARNTL Transcription Factors/physiology , Animals , Circadian Clocks/genetics , Circadian Clocks/immunology , Circadian Clocks/physiology , Circadian Rhythm/genetics , Cytokines/biosynthesis , Humans , Immunologic Memory , Mammals/genetics , Mammals/immunology , Mammals/physiology , Models, Immunological , Nutritional Physiological Phenomena , Sleep/immunology , Sleep/physiology
10.
Immunology ; 145(4): 583-96, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25939314

ABSTRACT

Dengue is a mosquito-borne disease that affects millions of people worldwide yearly. Currently, there is no vaccine or specific treatment available. Further investigation on dengue pathogenesis is required to better understand the disease and to identify potential therapeutic targets. The chemokine system has been implicated in dengue pathogenesis, although the specific role of chemokines and their receptors remains elusive. Here we describe the role of the CC-chemokine receptor CCR5 in Dengue virus (DENV-2) infection. In vitro experiments showed that CCR5 is a host factor required for DENV-2 replication in human and mouse macrophages. DENV-2 infection induces the expression of CCR5 ligands. Incubation with an antagonist prevents CCR5 activation and reduces DENV-2 positive-stranded (+) RNA inside macrophages. Using an immunocompetent mouse model of DENV-2 infection we found that CCR5(-/-) mice were resistant to lethal infection, presenting at least 100-fold reduction of viral load in target organs and significant reduction in disease severity. This phenotype was reproduced in wild-type mice treated with CCR5-blocking compounds. Therefore, CCR5 is a host factor required for DENV-2 replication and disease development. Targeting CCR5 might represent a therapeutic strategy for dengue fever. These data bring new insights on the association between viral infections and the chemokine receptor CCR5.


Subject(s)
Dengue Virus/physiology , Dengue/immunology , Macrophages/immunology , Receptors, CCR5/immunology , Virus Replication/immunology , Animals , Base Sequence , Dengue/drug therapy , Dengue/genetics , Humans , Macrophages/pathology , Macrophages/virology , Mice , Mice, Knockout , Molecular Sequence Data , Receptors, CCR5/genetics , Virus Replication/drug effects , Virus Replication/genetics
11.
Proc Natl Acad Sci U S A ; 112(23): 7231-6, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-25995365

ABSTRACT

The response to an innate immune challenge is conditioned by the time of day, but the molecular basis for this remains unclear. In myeloid cells, there is a temporal regulation to induction by lipopolysaccharide (LPS) of the proinflammatory microRNA miR-155 that correlates inversely with levels of BMAL1. BMAL1 in the myeloid lineage inhibits activation of NF-κB and miR-155 induction and protects mice from LPS-induced sepsis. Bmal1 has two miR-155-binding sites in its 3'-UTR, and, in response to LPS, miR-155 binds to these two target sites, leading to suppression of Bmal1 mRNA and protein in mice and humans. miR-155 deletion perturbs circadian function, gives rise to a shorter circadian day, and ablates the circadian effect on cytokine responses to LPS. Thus, the molecular clock controls miR-155 induction that can repress BMAL1 directly. This leads to an innate immune response that is variably responsive to challenges across the circadian day.


Subject(s)
ARNTL Transcription Factors/physiology , Circadian Rhythm , Immunity, Innate , Macrophages/immunology , MicroRNAs/physiology , 3' Untranslated Regions , ARNTL Transcription Factors/genetics , Adipose Tissue/metabolism , Animals , Cytokines/biosynthesis , Macrophages/metabolism , Mice , Mice, Knockout , NF-kappa B/metabolism
12.
PLoS One ; 10(3): e0118356, 2015.
Article in English | MEDLINE | ID: mdl-25793994

ABSTRACT

Rheumatoid Arthritis (RA) is a chronic disease characterized by persistent inflammation and pain. Alternative therapies to reduce these symptoms are needed. Marine algae are valuable sources of diverse bioactive compounds. Lithothamnion muelleri (Hapalidiaceae) is a marine algae with anti-inflammatory, antitumor, and immunomodulatory properties. Here, we investigated the potential anti-inflammatory and analgesic activities of L. muelleri in a murine model of antigen-induced arthritis (AIA) in mice. Our results demonstrate that treatment with L. muelleri prevented inflammation and hypernociception in arthritic mice. Mechanistically, the crude extract and the polysaccharide-rich fractions of L. muelleri may act impairing the production of the chemokines CXCL1 and CXCL2, and consequently inhibit neutrophil influx to the knee joint by dampening the adhesion step of leukocyte recruitment in the knee microvessels. Altogether our results suggest that treatment with L.muelleri has a potential therapeutic application in arthritis treatment.


Subject(s)
Arthritis, Experimental/pathology , Inflammation/pathology , Nociception , Rhodophyta/chemistry , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/drug therapy , Calcium Carbonate/chemistry , Cell Adhesion/drug effects , Endothelial Cells/drug effects , Endothelial Cells/pathology , Flow Cytometry , Joints/blood supply , Joints/drug effects , Joints/pathology , Leukocytes/drug effects , Leukocytes/pathology , Lymph Nodes/drug effects , Lymph Nodes/pathology , Male , Mice, Inbred C57BL , Nociception/drug effects , Polysaccharides/chemistry , Synovial Membrane/blood supply , Synovial Membrane/drug effects , Synovial Membrane/pathology
13.
J Immunol ; 193(4): 1911-9, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25015823

ABSTRACT

TLR4 interactor with leucine-rich repeats (TRIL) is a brain-enriched accessory protein that is important in TLR3 and TLR4 signaling. In this study, we generated Tril(-/-) mice and examined TLR responses in vitro and in vivo. We found a role for TRIL in both TLR4 and TLR3 signaling in mixed glial cells, consistent with the high level of expression of TRIL in these cells. We also found that TRIL is a modulator of the innate immune response to LPS challenge and Escherichia coli infection in vivo. Tril(-/-) mice produce lower levels of multiple proinflammatory cytokines and chemokines specifically within the brain after E. coli and LPS challenge. Collectively, these data uncover TRIL as a mediator of innate immune responses within the brain, where it enhances neuronal cytokine responses to infection.


Subject(s)
Brain/immunology , Carrier Proteins/immunology , Immunity, Innate/immunology , Membrane Proteins/immunology , Toll-Like Receptor 3/immunology , Toll-Like Receptor 4/immunology , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cells, Cultured , Chemokine CCL5/biosynthesis , Escherichia coli/immunology , Escherichia coli Infections/immunology , Intercellular Signaling Peptides and Proteins , Interleukin-6/biosynthesis , Lipopolysaccharides , Membrane Glycoproteins/immunology , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroglia/immunology , Poly I-C/pharmacology , Signal Transduction/immunology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 8/immunology , Tumor Necrosis Factor-alpha/biosynthesis
14.
Am J Pathol ; 184(7): 2023-34, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24952429

ABSTRACT

Irinotecan is a useful chemotherapeutic for the treatment of various cancers. Irinotecan treatment is associated with mucositis, which clearly limits the use of the drug. Mechanisms that account for mucositis are only partially known. This study assessed mechanisms and the role of inflammasome activation in irinotecan-induced mucositis. Mucositis in mice was induced by irinotecan injection in C57BL/6 wild-type, gp91phox(-/-), il-18(-/-), casp-1(-/-), and asc(-/-) mice once a day for 4 consecutive days. In some experiments, mice received apocynin to inhibit NADPH oxidase (NOX), IL-1 receptor antagonist, or IL-18 binding protein to prevent activation of IL-1 and IL-18 receptors, respectively. Mice were euthanized 7 days after the beginning of irinotecan treatment, and small intestines were collected for analysis. Irinotecan treatment resulted in increased IL-1ß and IL-18 production in ileum and NOX-2-dependent oxidative stress. gp91phox(-/-) and apocynin-treated mice had diminished oxidative stress and less severe mucositis. Furthermore, treatment with apocynin decreased caspase-1 activation and IL-1ß and IL-18 production in the ileum. asc(-/-) and casp-1(-/-) mice also had less intestinal injury and decreased IL-1ß and IL-18 production. Finally, both the absence of IL-18 and IL-1ß resulted in reduced inflammatory response and attenuated intestinal injury. NOX-2-derived oxidative stress mediates inflammasome activation and inflammasome-dependent production of IL-1ß and IL-18, which mediate tissue injury during irinotecan-induced mucositis in mice.


Subject(s)
Inflammasomes/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Mucositis/metabolism , Reactive Oxygen Species/metabolism , Animals , Camptothecin/adverse effects , Camptothecin/analogs & derivatives , Caspase 1/metabolism , Ileum/metabolism , Ileum/pathology , Irinotecan , Male , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mucositis/chemically induced , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Oxidative Stress
15.
Med Microbiol Immunol ; 203(4): 231-50, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24723052

ABSTRACT

Dengue is a mosquito-borne disease caused by one of four serotypes of Dengue virus (DENV-1-4). Epidemiologic and observational studies demonstrate that the majority of severe dengue cases, dengue hemorrhagic fever and dengue shock syndrome (DHF/DSS), occurs predominantly in either individuals with cross-reactive immunity following a secondary heterologous infection or in infants with primary DENV infections born from dengue-immune mothers, suggesting that B-cell-mediated and antibody responses impact on disease evolution. We demonstrate here that B cells play a pivotal role in host responses against primary DENV infection in mice. After infection, µMT(-/-) mice showed increased viral loads followed by severe disease manifestation characterized by intense thrombocytopenia, hemoconcentration, cytokine production and massive liver damage that culminated in death. In addition, we show that poly and monoclonal anti-DENV-specific antibodies can sufficiently increase viral replication through a suppression of early innate antiviral responses and enhance disease manifestation, so that a mostly non-lethal illness becomes a fatal disease resembling human DHF/DSS. Finally, treatment with intravenous immunoglobulin containing anti-DENV antibodies confirmed the potential enhancing capacity of subneutralizing antibodies to mediate virus infection and replication and induce severe disease manifestation of DENV-infected mice. Thus, our results show that humoral responses unleashed during DENV infections can exert protective or pathological outcomes and provide insight into the pathogenesis of this important human pathogen.


Subject(s)
Antibody-Dependent Enhancement , Dengue Virus/immunology , Dengue/immunology , Dengue/pathology , Immunity, Innate , Animals , B-Lymphocytes/immunology , Cytokines/blood , Death , Liver/pathology , Mice, Inbred C57BL , Mice, Knockout , Thrombocytopenia , Viral Load
16.
Cell Res ; 23(9): 1065-6, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23774266

ABSTRACT

A cyclic dinucleotide comprised of GMP and AMP was previously shown to be a key intermediate during activation of innate immune responses to cytosolic DNA. A report by Patel and Tuschl groups published in Cell reveals the structure of the enzyme involved in the synthesis of this second messenger and identifies this cyclic dinucleotide as a unique compound in metazoan cell signaling.


Subject(s)
Dinucleoside Phosphates/metabolism , Nucleotides, Cyclic/metabolism , Nucleotidyltransferases/chemistry , Second Messenger Systems , Animals , Humans
17.
Eur J Immunol ; 43(6): 1529-44, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23505056

ABSTRACT

Dengue virus (DENV), a mosquito-borne flavivirus, is a public health problem in many tropical countries. IL-22 and IL-17A are key cytokines in several infectious and inflammatory diseases. We have assessed the contribution of IL-22 and IL-17A in the pathogenesis of experimental dengue infection using a mouse-adapted DENV serotype 2 strain (P23085) that causes a disease that resembles severe dengue in humans. We show that IL-22 and IL-17A are produced upon DENV-2 infection in immune-competent mice. Infected IL-22(-/-) mice had increased lethality, neutrophil accumulation and pro-inflammatory cytokines in tissues, notably IL-17A. Viral load was increased in spleen and liver of infected IL-22(-/-) mice. There was also more severe liver injury, as seen by increased transaminases levels and tissue histopathology. γδ T cells and NK cells are sources of IL-17A and IL-22, respectively, in liver and spleen. We also show that DENV-infected HepG2 cells treated with rhIL-22 had reduced cell death and decreased IL-6 production. IL-17RA(-/-) mice were protected upon infection and IL-17A-neutralizing-Ab-treatment partially reversed the phenotype observed in IL-22(-/-) -infected mice. We suggest that disrupting the balance between IL-22 and IL-17A levels may represent an important strategy to reduce inflammation and tissue injury associated with severe dengue infection.


Subject(s)
Dengue Virus/immunology , Dengue/immunology , Inflammation Mediators/metabolism , Interleukin-17/metabolism , Interleukins/metabolism , Liver/metabolism , Neutrophils/immunology , Animals , Apoptosis/drug effects , Disease Models, Animal , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Hep G2 Cells , Humans , Inflammation/genetics , Interleukin-17/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukins/genetics , Interleukins/immunology , Liver/immunology , Liver/pathology , Liver/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/virology , Receptors, Interleukin-17/genetics , Viral Load/genetics , Interleukin-22
18.
PLoS Negl Trop Dis ; 6(5): e1663, 2012.
Article in English | MEDLINE | ID: mdl-22666512

ABSTRACT

There are few animal models of dengue infection, especially in immunocompetent mice. Here, we describe alterations found in adult immunocompetent mice inoculated with an adapted Dengue virus (DENV-3) strain. Infection of mice with the adapted DENV-3 caused inoculum-dependent lethality that was preceded by several hematological and biochemical changes and increased virus dissemination, features consistent with severe disease manifestation in humans. IFN-γ expression increased after DENV-3 infection of WT mice and this was preceded by increase in expression of IL-12 and IL-18. In DENV-3-inoculated IFN-γ(-/-) mice, there was enhanced lethality, which was preceded by severe disease manifestation and virus replication. Lack of IFN-γ production was associated with diminished NO-synthase 2 (NOS2) expression and higher susceptibility of NOS2(-/-) mice to DENV-3 infection. Therefore, mechanisms of protection to DENV-3 infection rely on IFN-γ-NOS2-NO-dependent control of viral replication and of disease severity, a pathway showed to be relevant for resistance to DENV infection in other experimental and clinical settings. Thus, the model of DENV-3 infection in immunocompetent mice described here represents a significant advance in animal models of severe dengue disease and may provide an important tool to the elucidation of immunopathogenesis of disease and of protective mechanisms associated with infection.


Subject(s)
Dengue Virus/immunology , Dengue Virus/pathogenicity , Dengue/immunology , Dengue/pathology , Disease Models, Animal , Interferon-gamma/immunology , Adaptation, Biological , Animals , Cytokines/metabolism , Dengue/mortality , Dengue/virology , Interferon-gamma/deficiency , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/deficiency , Nitric Oxide Synthase Type II/immunology , Survival Analysis
19.
Br J Nutr ; 108(10): 1829-38, 2012 Nov 28.
Article in English | MEDLINE | ID: mdl-22273003

ABSTRACT

Rheumatoid arthritis (RA) is a chronic inflammatory disease that mainly targets the synovial membrane, cartilage and bone. It affects 1 % of the population and is associated with significant morbidity and increased mortality. Se is an essential trace element with antioxidant properties and the ability to modulate the immune responses. Selemax® is an inactive yeast (Saccharomyces cerevisiae) enriched with organic Se. The aim of the present study was to investigate the effects of Selemax® administration in models of an antigen-induced arthritis (AIA) in C57BL/6 mice, and of an adjuvant-induced arthritis (AdIA) in Holtzman rats. As control, the animals were treated with the same inactivated yeast species that was not enriched for Se. In the AIA model, treatment with different doses of Selemax® (0·01, 0·1, 1 and 10 % added to food) significantly decreased the number of inflammatory cells recruited to the knee cavity, essentially by reducing the number of neutrophils. Levels of proinflammatory cytokines, including TNF-α, IL-1ß and chemokine (C-X-C motif) ligand 1/keratinocyte chemoattractant (CXCL1/KC), were also reduced in the peri-articular tissue of mice treated with Selemax® at the tested dose (1 %). In the AdIA model in rats, Selemax® treatment decreased paw oedema and hypernociception. This reduction was associated with inhibition of the influx of proinflammatory cells. Therefore, treatment with Selemax® is associated with amelioration of several inflammatory and functional parameters in models of arthritis, suggesting that this Se-enriched yeast should be evaluated further in patients with RA.


Subject(s)
Arthritis/chemically induced , Arthritis/drug therapy , Dietary Supplements , Selenium/administration & dosage , Selenium/therapeutic use , Animals , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression Regulation/drug effects , Macrophages/drug effects , Male , Metalloporphyrins , Mice , Mice, Inbred C57BL , Neutrophils , Peroxidase/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species , Serum Albumin, Bovine/toxicity , Yeasts
20.
J Immunol ; 188(3): 1411-20, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22210917

ABSTRACT

Mammals are colonized by an astronomical number of commensal microorganisms on their environmental exposed surfaces. These symbiotic species build up a complex community that aids their hosts in several physiological activities. We have shown that lack of intestinal microbiota is accompanied by a state of active IL-10-mediated inflammatory hyporesponsiveness. The present study investigated whether the germfree state and its hyporesponsive phenotype alter host resistance to an infectious bacterial insult. Experiments performed in germfree mice infected with Klebsiella pneumoniae showed that these animals are drastically susceptible to bacterial infection in an IL-10-dependent manner. In germfree mice, IL-10 restrains proinflammatory mediator production and neutrophil recruitment and favors pathogen growth and dissemination. Germfree mice were resistant to LPS treatment. However, priming of these animals with several TLR agonists recovered their inflammatory responsiveness to sterile injury. LPS pretreatment also rendered germfree mice resistant to pulmonary K. pneumoniae infection, abrogated IL-10 production, and restored TNF-α and CXCL1 production and neutrophil mobilization into lungs of infected germfree mice. This effective inflammatory response mounted by LPS-treated germfree mice resulted in bacterial clearance and enhanced survival upon infection. Therefore, host colonization by indigenous microbiota alters the way the host reacts to environmental infectious stimuli, probably through activation of TLR-dependent pathways. Symbiotic gut colonization enables proper inflammatory response to harmful insults to the host, and increases resilience of the entire mammal-microbiota consortium to environmental pressures.


Subject(s)
Inflammation/etiology , Klebsiella Infections/immunology , Toll-Like Receptors/metabolism , Animals , Germ-Free Life , Host-Pathogen Interactions/immunology , Interleukin-10/immunology , Intestines/immunology , Intestines/microbiology , Klebsiella pneumoniae , Metagenome/immunology , Mice , Toll-Like Receptors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...