Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters











Publication year range
1.
FASEB J ; 33(10): 10794-10807, 2019 10.
Article in English | MEDLINE | ID: mdl-31284755

ABSTRACT

Visceral leishmaniasis is a deadly illness caused by Leishmania donovani that provokes liver and spleen inflammation and tissue destruction. In cutaneous leishmaniasis, the protein of L. major, named inhibitor of serine peptidases (ISP) 2, inactivates neutrophil elastase (NE) present at the macrophage surface, resulting in blockade of TLR4 activation, prevention of TNF-α and IFN-ß production, and parasite survival. We report poor intracellular growth of L. donovani in macrophages from knockout mice for NE (ela-/-), TLR4, or TLR2. NE and TLR4 colocalized with the parasite in the parasitophorous vacuole. Parasite load in the liver and spleen of ela-/- mice were reduced and accompanied by increased NO and decreased TGF-ß production. Expression of ISP2 was not detected in L. donovani, and a transgenic line constitutively expressing ISP2, displayed poor intracellular growth in macrophages and decreased burden in mice. Infected ela-/- macrophages displayed significantly lower IFN-ß mRNA than background mice macrophages, and the intracellular growth was fully restored by exogenous IFN-ß. We propose that L. donovani utilizes the host NE-TLR machinery to induce IFN-ß necessary for parasite survival and growth during early infection. Low or absent expression of parasite ISP2 in L. donovani is necessary to preserve the activation of the NE-TLR pathway.-Dias, B. T., Dias-Teixeira, K. L., Godinho, J. P., Faria, M. S., Calegari-Silva, T., Mukhtar, M. M., Lopes, U. G., Mottram, J. C., Lima, A. P. C. A. Neutrophil elastase promotes Leishmania donovani infection via interferon-ß.


Subject(s)
Interferon-beta/metabolism , Leishmania donovani/pathogenicity , Leishmaniasis, Visceral/etiology , Leukocyte Elastase/metabolism , Animals , Animals, Genetically Modified , Leishmania donovani/genetics , Leishmania donovani/physiology , Leishmaniasis, Visceral/metabolism , Leishmaniasis, Visceral/parasitology , Leukocyte Elastase/deficiency , Leukocyte Elastase/genetics , Macrophages/metabolism , Macrophages/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protozoan Proteins/genetics , Protozoan Proteins/physiology , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
2.
Am J Physiol Renal Physiol ; 315(3): F460-F468, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29717937

ABSTRACT

Interstitial Cystitis/Bladder Pain Syndrome (IC/BPS) is a chronic inflammatory disease without consistently effective treatment. We investigate the role of toll-like receptor 4 (TLR4) on voiding dysfunction and inflammation in the cyclophosphamide (CYP)-induced mouse cystitis. Male C57BL/6 [wild-type, (WT)] and/or TLR4 knockout (TLR4-/-) mice were treated with an injection of CYP (300 mg/kg, 24 h) or saline (10 ml/kg). The pharmacological blockade of the TLR4 by resatorvid (10 mg/kg) was also performed 1 h prior CYP-injection in WT mice. Urodynamic profiles were assessed by voiding stain on filter paper and filling cystometry. Contractile responses to carbachol were measured in isolated bladders. In CYP-exposed WT mice, mRNA for TLR4, myeloid differentiation primary response 88, and TIR-domain-containing adapter-inducing interferon-ß increased by 45%, 72%, and 38%, respectively ( P < 0.05). In free-moving mice, CYP-exposed mice exhibited a higher number of urinary spots and smaller urinary volumes. Increases of micturition frequency and nonvoiding contractions, concomitant with decreases of intercontraction intervals and capacity, were observed in the filling cystometry of WT mice ( P < 0.05). Carbachol-induced bladder contractions were significantly reduced in the CYP group, which was paralleled by reduced mRNA for M2 and M3 muscarinic receptors. These functional and molecular alterations induced by CYP were prevented in TLR4-/- and resatorvid-treated mice. Additionally, the increased levels of inflammatory markers induced by CYP exposure, myeloperoxidase activity, interleukin-6, and tumor necrosis factor-alpha were significantly reduced by resatorvid treatment. Our findings reveal a central role for the TLR4 signaling pathway in initiating CYP-induced bladder dysfunction and inflammation and thus emphasize that TLR4 receptor blockade may have clinical value for IC/BPS treatment.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cyclophosphamide , Cystitis, Interstitial/prevention & control , Sulfonamides/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/deficiency , Urinary Bladder/drug effects , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cystitis, Interstitial/chemically induced , Cystitis, Interstitial/genetics , Cystitis, Interstitial/metabolism , Disease Models, Animal , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Peroxidase/metabolism , Receptor, Muscarinic M2/genetics , Receptor, Muscarinic M2/metabolism , Receptor, Muscarinic M3/genetics , Receptor, Muscarinic M3/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/metabolism , Urinary Bladder/metabolism , Urinary Bladder/physiopathology , Urination/drug effects , Urodynamics/drug effects
3.
Glia ; 65(7): 1137-1151, 2017 07.
Article in English | MEDLINE | ID: mdl-28398652

ABSTRACT

Inflammation has long been implicated as a contributor to pathogenesis in neurobrucellosis. Many of the associated neurocognitive symptoms of neurobrucellosis may be the result of neuronal dysfunction resulting from the inflammatory response induced by Brucella abortus infection in the central nervous system. In this manuscript, we describe an immune mechanism for inflammatory activation of microglia that leads to neuronal death upon B. abortus infection. B. abortus was unable to infect or harm primary cultures of mouse neurons. However, when neurons were co-cultured with microglia and infected with B. abortus significant neuronal loss occurred. This phenomenon was dependent on TLR2 activation by Brucella lipoproteins. Neuronal death was not due to apoptosis, but it was dependent on the microglial release of nitric oxide (NO). B. abortus infection stimulated microglial proliferation, phagocytic activity and engulfment of neurons. NO secreted by B. abortus-activated microglia induced neuronal exposure of the "eat-me" signal phosphatidylserine (PS). Blocking of PS-binding to protein milk fat globule epidermal growth factor-8 (MFG-E8) or microglial vitronectin receptor-MFG-E8 interaction was sufficient to prevent neuronal loss by inhibiting microglial phagocytosis without affecting their activation. Taken together, our results indicate that B. abortus is not directly toxic to neurons; rather, these cells become distressed and are killed by phagocytosis in the inflammatory surroundings generated by infected microglia. Neuronal loss induced by B. abortus-activated microglia may explain, in part, the neurological deficits observed during neurobrucellosis.


Subject(s)
Brucella abortus/pathogenicity , Cell Death/physiology , Inflammation/metabolism , Microglia/microbiology , Microglia/physiology , Neurons/pathology , Phagocytosis/physiology , Animals , Antigens, Bacterial/toxicity , Bacterial Outer Membrane Proteins/toxicity , Cell Death/genetics , Cells, Cultured , Embryo, Mammalian , Gene Expression Regulation, Bacterial/physiology , Inflammation/chemically induced , Inflammation/pathology , Lipopolysaccharides/pharmacology , Lipoproteins/metabolism , Lipoproteins/toxicity , Mice , Mice, Inbred BALB C , Mice, Transgenic , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Neurons/cytology , Neurons/drug effects , Nitric Oxide/metabolism , Prosencephalon/cytology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
4.
Histochem Cell Biol ; 148(1): 49-60, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28293722

ABSTRACT

Tissue damage triggers innate immune response mediated by Toll-like receptor 4 (TLR) that recognizes endogenous host danger molecules associated with cell death and tissue inflammation, although the precise role of TLR-4 signaling in muscle tissue repair is still uncertain. Previously, we observed that TLR-4 exerted a protective effect preventing excessive muscular damage induced by Bothrops jararacussu crude venom. This study aimed to evaluate the involvement of TLR-4 at early stages of muscular tissue remodeling in distinct mouse strains after injection of purified snake venom. Muscular injury was induced by injection of 25 µl (0.05 mg/ml) of cardiotoxin (CTX) from Naja mossambica in the gastrocnemius muscle of C3H/HeN (wild-type); C3H/HeJ mice that express a non-functional TLR-4 receptor, C57BL/6 and Tlr4 -/- (B6 background) mice. Comparing to control, Tlr4 -/- mice presented at early stages (3 DPI) of muscle injury mild inflammation with low MMP-9 activity, scarce macrophage infiltration and premature change to anti-inflammatory phenotype, low TNF-α mRNA levels and reduced myogenin expression, with low regeneration and tissue remodeling. The presence of more Ly6Cneg macrophages in Tlr4 -/- mice at 3 DPI indicates that TLR-4 may influence the differentiation into Ly6Cneg or likely affect proliferation of such cells in the muscle. The present study shows that TLR-4 deficiency and genetic background influence the outcome of muscular tissue repair in aseptic lesions and yet still maintaining some level of signaling in the TLR4-mutant mice.


Subject(s)
Cardiotoxins/pharmacology , Muscle Fibers, Skeletal/drug effects , Toll-Like Receptor 4/deficiency , Animals , Cardiotoxins/administration & dosage , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Toll-Like Receptor 4/metabolism
5.
J Microbiol ; 54(11): 745-752, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27796929

ABSTRACT

Using clonal phylogenetic methods, it has been demonstrated that O111:H25 atypical enteropathogenic E. coli (aEPEC) strains belong to distinct clones, suggesting the possibility that their ability to interact with different hosts and abiotic surfaces can vary from one clone to another. Accordingly, the ability of O111:H25 aEPEC strains derived from human, cat and dogs to adhere to epithelial cells has been investigated, along with their ability to interact with macrophages and to form biofilms on polystyrene, a polymer used to make biomedical devices. The results demonstrated that all the strains analyzed were able to adhere to, and to form pedestals on, epithelial cells, mechanisms used by E. coli to become strongly attached to the host. The strains also show a Localized-Adherence-Like (LAL) pattern of adhesion on HEp-2 cells, a behavior associated with acute infantile diarrhea. In addition, the O111:H25 aEPEC strains derived either from human or domestic animals were able to form long filaments, a phenomenon used by some bacteria to avoid phagocytosis. O111:H25 aEPEC strains were also encountered inside vacuoles, a characteristic described for several bacterial strains as a way of protecting themselves against the environment. They were also able to induce TNF-α release via two routes, one dependent on TLR-4 and the other dependent on binding of Type I fimbriae. These O111:H25 strains were also able to form biofilms on polystyrene. In summary the results suggest that, regardless of their source (i.e. linked to human origin or otherwise), O111:H25 aEPEC strains carry the potential to cause human disease.


Subject(s)
Bacterial Adhesion , Enteropathogenic Escherichia coli/metabolism , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Animals , Biofilms/growth & development , Cats , Dogs , Enteropathogenic Escherichia coli/isolation & purification , Enteropathogenic Escherichia coli/ultrastructure , Epithelial Cells/microbiology , Escherichia coli Proteins , Fimbriae, Bacterial/immunology , Humans , Macrophages/immunology , Macrophages/microbiology , Mice , Phylogeny , Polystyrenes , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/metabolism , Virulence Factors
6.
PLoS One ; 10(10): e0139350, 2015.
Article in English | MEDLINE | ID: mdl-26448184

ABSTRACT

We investigated whether the pathways linked to Toll-like receptors 2 and 4 (TLRs) are involved in renal ischemia-reperfusion (I/R)-induced cardiac hypertrophy. Wild type (WT) C57BL/6J, TLR2-/- and TLR4-/- mice were subjected to left kidney ischemia for 60 min followed by reperfusion for 5, 8, 12 and 15 days. Proton density magnetic resonance showed alterations in the injured kidney from WT mice, together with signs of parenchymal edema and higher levels of vimentin mRNA, accompanied by: (i) small, but significant, increase in serum urea after 24 h, (ii) 100% increase in serum creatinine at 24 h. A serum peak of inflammatory cytokines occurred after 5 days of reperfusion. Heart weight/body weight and heart weight/tibia length ratios increased after 12 and 15 days of reperfusion, respectively. Cardiac hypertrophy markers, B-type natriuretic peptide (BNP) and α-actin, left ventricle mass, cardiac wall thickness and myocyte width increased after 15 days of reperfusion, together with longer QTc and action potential duration. Cardiac TLRs, MyD88, HSP60 and HSP70 mRNA levels also increased. After 15 days of reperfusion, absence of TLRs prevented cardiac hypertrophy, as reflected by similar values of left ventricular cardiac mass and heart weight/body weight ratio compared to the transgenic Sham. Renal tissular injury also ameliorated in both knockout mice, as revealed by the comparison of their vimentin mRNA levels with those found in the WT on the same day after I/R. The I/R TLR2-/- group had TNF-α, IFN-γ and IL-1ß levels similar to the non-I/R group, whereas the TLR4-/- group conserved the p-NF-κB/NF- κB ratio contrasting with that found in TLR2-/-. We conclude: (i) TLRs are involved in renal I/R-induced cardiac hypertrophy; (ii) absence of TLRs prevents I/R-induced cardiac hypertrophy, despite renal lesions seeming to evolve towards those of chronic disease; (iii) TLR2 and TLR4 selectively regulate the systemic inflammatory profile and NF- κB activation.


Subject(s)
Cardiomegaly/etiology , Ischemia/complications , Reperfusion Injury/complications , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Actins/metabolism , Animals , Cardiomegaly/metabolism , Cardiomegaly/prevention & control , Chaperonin 60/genetics , Chaperonin 60/metabolism , Cytokines/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heart/physiology , Kidney/physiopathology , Kidney Diseases/complications , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Myocardium/metabolism , Natriuretic Peptide, Brain/metabolism , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 4/deficiency , Vimentin/genetics , Vimentin/metabolism
7.
PLoS One ; 10(5): e0126827, 2015.
Article in English | MEDLINE | ID: mdl-25973756

ABSTRACT

Brucella Lumazine Synthase (BLS) is a highly immunogenic decameric protein which can accept the fusion of foreign proteins at its ten N-termini. These chimeras are very efficient to elicit systemic and oral immunity without adjuvants. BLS signaling via Toll-Like Receptor 4 (TLR4) regulates innate and adaptive immune responses, inducing dendritic cell maturation and CD8(+) T-cell cytotoxicity. In this work we study the effect induced by BLS in TLR4-expressing B16 melanoma. In order to evaluate the effectiveness of BLS as a preventive vaccine, C57BL/6J mice were immunized with BLS or BLS-OVA, and 35 days later were subcutaneously inoculated with B16-OVA melanoma. BLS or BLS-OVA induced a significant inhibition of tumor growth, and 50% of mice immunized with the highest dose of BLS did not develop visible tumors. This effect was not observed in TLR4-deficient mice. For treatment experiments, mice were injected with BLS or BLS-OVA 2 days after the inoculation of B16 cells. Both treatments induced significant and equal tumor growth delay and increased survival. Moreover, BLS and BLS-OVA stimulation were also effective in TLR4-deficient mice. In order to study whether BLS has a direct effect on tumor cells, B16 cells were preincubated with BLS, and after 48h, cells were inoculated. Tumors induced by BLS-stimulated cells had inhibited growth and survival was increased. In the BLS group, 40% of mice did not develop tumors. This effect was abolished by the addition of TLR4/MD2 blocking antibody to cells before BLS stimulation. Our work demonstrates that BLS immunization induces a preventive antitumor response that depends on mice TLR4. We also show that BLS generates a therapeutic effect in mice inoculated with B16 cells. Our results show that BLS acts directly in cultured tumor cells via TLR4, highly suggesting that BLS elicits its therapeutic effects acting on the TLR4 from B16 melanoma cells.


Subject(s)
Brucella/enzymology , Multienzyme Complexes/metabolism , Toll-Like Receptor 4/genetics , Animals , Apoptosis , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Melanoma, Experimental/drug therapy , Melanoma, Experimental/mortality , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Multienzyme Complexes/genetics , Multienzyme Complexes/immunology , Ovalbumin/genetics , Ovalbumin/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Survival Rate , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/metabolism , Transplantation, Homologous
8.
Hepatology ; 60(3): 990-1002, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24798499

ABSTRACT

UNLABELLED: Acetaminophen (APAP) toxicity is the most common cause of acute liver failure in industrialized countries. Understanding the mechanisms of APAP-induced liver injury as well as other forms of sterile liver injury is critical to improve the care of patients. Recent studies demonstrate that danger signaling and inflammasome activation play a role in APAP-induced injury. The aim of these investigations was to test the hypothesis that benzyl alcohol (BA) is a therapeutic agent that protects against APAP-induced liver injury by modulation of danger signaling. APAP-induced liver injury was dependent, in part, on Toll-like receptor (TLR)9 and receptor for advanced glycation endproducts (RAGE) signaling. BA limited liver injury over a dose range of 135-540 µg/g body weight or when delivered as a pre-, concurrent, or post-APAP therapeutic. Furthermore, BA abrogated APAP-induced cytokines and chemokines as well as high-mobility group box 1 release. Moreover, BA prevented APAP-induced inflammasome signaling as determined by interleukin (IL)-1ß, IL-18, and caspase-1 cleavage in liver tissues. Interestingly, the protective effects of BA on limiting liver injury and inflammasome activation were dependent on TLR4 signaling, but not TLR2 or CD14. Cell-type-specific knockouts of TLR4 were utilized to further determine the protective mechanisms of BA. These studies found that TLR4 expression specifically in myeloid cells (LyzCre-tlr4-/-) were necessary for the protective effects of BA. CONCLUSION: BA protects against APAP-induced acute liver injury and reduced inflammasome activation in a TLR4-dependent manner. BA may prove to be a useful adjunct in the treatment of APAP and other forms of sterile liver injury.


Subject(s)
Benzyl Alcohol/therapeutic use , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Toll-Like Receptor 4/physiology , Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Animals , Cells, Cultured , Chemical and Drug Induced Liver Injury/etiology , HMGB1 Protein/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Random Allocation , Receptor for Advanced Glycation End Products , Receptors, Immunologic/physiology , Toll-Like Receptor 4/deficiency
9.
Circ Res ; 114(10): 1596-600, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24637196

ABSTRACT

RATIONALE: Recent studies demonstrate a role for toll-like receptor 4 (TLR4) in the pathogenesis of pulmonary hypertension (PH); however, the cell types involved in mediating the effects of TLR4 remain unknown. OBJECTIVES: The objective of this study was to determine the contribution of TLR4 expressed on nonparenchymal cells to the pathogenesis of PH. METHODS AND RESULTS: TLR4 bone marrow chimeric mice revealed an equal contribution of TLR4 on nonparenchymal and parenchymal cells in the pathogenesis of PH as determined by measuring right ventricular (RV) systolic pressure and RV hypertrophy. However, the deletion of TLR4 from myeloid lineage cells had no effect on the development of PH because we found no difference in RV systolic pressure or RV hypertrophy in wild-type versus LysM-TLR4(-/-) mice. To explore the potential role of platelet TLR4 in the pathogenesis of PH, platelet-specific TLR4(-/-) mice were generated (PF4-TLR4(-/-) mice). TLR4(-/-) platelets from either global TLR4(-/-) or PF4-TLR4(-/-) mice were functional but failed to respond to lipopolysaccharide, demonstrating a lack of TLR4. PF4-TLR4(-/-) mice demonstrated significant protection from hypoxia-induced PH, including attenuated increases in RV systolic pressure and RV hypertrophy, decreased platelet activation, and less pulmonary vascular remodeling. The deletion of TLR4 from platelets attenuated serotonin release after chronic hypoxia, and lipopolysaccharide-stimulated platelets released serotonin and promoted pulmonary artery smooth muscle cell proliferation in a serotonin-dependent manner. CONCLUSIONS: Our data demonstrate that TLR4 on platelets contributes to the pathogenesis of PH and further highlights the role of platelets in PH.


Subject(s)
Blood Platelets/pathology , Gene Deletion , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/prevention & control , Toll-Like Receptor 4/deficiency , Animals , Blood Platelets/metabolism , Coculture Techniques , Humans , Hypertension, Pulmonary/blood , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiology , Radiation Chimera , Toll-Like Receptor 4/blood , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/physiology
10.
Circulation ; 128(11 Suppl 1): S152-6, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-24030400

ABSTRACT

BACKGROUND: Paraplegia continues to complicate thoracoabdominal aortic interventions. The elusive mechanism of spinal cord ischemia-reperfusion injury has delayed the development of pharmacological adjuncts. Microglia, the resident macrophages of the central nervous system, can have pathological responses after a variety of insults. This can occur through toll-like receptor 4 (TLR-4) in stroke models. We hypothesize that spinal cord ischemia-reperfusion injury after aortic occlusion results from TLR-4-mediated microglial activation in mice. METHODS AND RESULTS: TLR-4 mutant and wild-type mice underwent aortic occlusion for 5 minutes, followed by 60 hours of reperfusion when spinal cords were removed for analysis. Spinal cord cytokine production and microglial activation were assessed at 6 and 36 hours after surgery. Isolated microglia from mutant and wild-type mice were subjected to oxygen and glucose deprivation for 24 hours, after which the expression of TLR-4 and proinflammatory cytokines was analyzed. Mice without functional TLR-4 demonstrated decreased microglial activation and cytokine production and had preserved functional outcomes and neuronal viability after thoracic aortic occlusion. After oxygen and glucose deprivation, wild-type microglia had increased TLR-4 expression and production of proinflammatory cytokines. CONCLUSIONS: The absence of functional TLR-4 attenuated neuronal injury and microglial activation after thoracic aortic occlusion in mice. Furthermore, microglial upregulation of TLR-4 occurred after oxygen and glucose deprivation, and the absence of functional TLR-4 significantly attenuated the production of proinflammatory cytokines. In conclusion, TLR-4-mediated microglia activation in the spinal cord after aortic occlusion is critical in the mechanism of paraplegia after aortic cross-clamping and may provide targets for pharmacological intervention.


Subject(s)
Microglia/metabolism , Reperfusion Injury/metabolism , Spinal Cord Ischemia/metabolism , Toll-Like Receptor 4/physiology , Animals , Cell Survival/physiology , Cells, Cultured , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C3H , Mice, Knockout , Reperfusion Injury/pathology , Spinal Cord Ischemia/pathology , Toll-Like Receptor 4/deficiency
11.
Vaccine ; 31(41): 4528-35, 2013 Sep 23.
Article in English | MEDLINE | ID: mdl-23933366

ABSTRACT

Virulent strains of Rhodococcus equi have a large plasmid of 80-90kb, which encodes several virulence-associated proteins (Vap), including VapA, a lipoprotein highly associated with disease. We have previously demonstrated that oral immunisation with attenuated Salmonella enterica Typhimurium strain expressing the antigen VapA (STM VapA+) induces specific and long-term humoral and cellular immunity against R. equi. It was shown that VapA activates Toll-like receptor 2 (TLR2) on macrophages by establishing an interaction that ultimately favours immunity against R. equi infection. The purpose of this study was to evaluate the immune response triggered by nasal immunisation with STM VapA+ and to determine whether TLR2 supports the vaccine effect. We developed an optimised protocol for a single nasal immunisation that conferred protection against R. equi infection in mice, which was manifested by efficient R. equi clearance in challenged animals. Nasal vaccination with STM VapA+ has also induced protection in Tlr2(-/-) mice and mice with non-functional TLR4. Moreover, spleen cells of vaccinated mice augmented T-bet expression, as well as the production of IL-12, IFN-γ, nitric oxide and hydrogen peroxide. Notably, the population of CD4(+) T cells with memory phenotype significantly increased in the spleens of vaccinated mice challenged 1 or 5 months after immunisation. In these animals, the spleen bacterial burden was also reduced. When similar experimental procedures were performed in TLR2 knockout mice, an increase in CD4(+) T cells with memory phenotype was not observed. Consequently, we conclude that nasal vaccination with attenuated Salmonella expressing the R. equi virulence factor VapA confers long-lasting protection against experimental rhodoccocosis and that TLR2 engagement was not crucial to induce this protection but may be required for a long-term immune response.


Subject(s)
Actinomycetales Infections/prevention & control , Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Drug Carriers , Rhodococcus equi/immunology , Salmonella typhimurium/genetics , Toll-Like Receptor 2/immunology , Actinomycetales Infections/immunology , Administration, Intranasal , Animals , Bacterial Load , Bacterial Proteins/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Female , Genetic Vectors , Immunologic Memory , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Rhodococcus equi/genetics , Spleen/immunology , Spleen/microbiology , T-Lymphocytes/immunology , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/immunology
12.
Hepatology ; 58(1): 374-87, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23460269

ABSTRACT

UNLABELLED: Ischemia-reperfusion (I/R) injury is a process whereby an initial hypoxic insult and subsequent return of blood flow leads to the propagation of innate immune responses and organ injury. The necessity of the pattern recognition receptor, Toll-like receptor (TLR)4, for this innate immune response has been previously shown. However, TLR4 is present on various cell types of the liver, both immune and nonimmune cells. Therefore, we sought to determine the role of TLR4 in individual cell populations, specifically, parenchymal hepatocytes (HCs), myeloid cells, including Kupffer cells, and dendritic cells (DCs) subsequent to hepatic I/R. When HC-specific (Alb-TLR4(-/-) ) and myeloid-cell-specific (Lyz-TLR4(-/-) ) TLR4 knockout (KO) mice were subjected to warm hepatic ischemia, there was significant protection in these mice, compared to wild type (WT). However, the protection afforded in these two strains was significantly less than global TLR4 KO (TLR4(-/-) ) mice. DC-specific TLR4(-/-) (CD11c-TLR4(-/-) ) mice had significantly increased hepatocellular damage, compared to WT mice. Circulating levels of high-mobility group box 1 (HMGB1) were significantly reduced in Alb-TLR4(-/-) mice, compared to WT, Lyz-TLR4(-/-) , CD11c-TLR4(-/-) mice and equivalent to global TLR4(-/-) mice, suggesting that TLR4-mediated HMGB1 release from HCs may be a source of HMGB1 after I/R. HCs exposed to hypoxia responded by rapidly phosphorylating the mitogen-activated protein kinases, c-Jun-N-terminal kinase (JNK) and p38, in a TLR4-dependent manner; inhibition of JNK decreased release of HMGB1 after both hypoxia in vitro and I/R in vivo. CONCLUSION: These results provide insight into the individual cellular response of TLR4. The parenchymal HC is an active participant in sterile inflammatory response after I/R through TLR4-mediated activation of proinflammatory signaling and release of danger signals, such as HMGB1.


Subject(s)
Hepatocytes/immunology , Immunity, Innate/physiology , Liver/immunology , Reperfusion Injury/immunology , Toll-Like Receptor 4/physiology , Animals , Dendritic Cells/immunology , HMGB1 Protein/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Kupffer Cells/immunology , Male , Mice , Mice, Knockout , Myeloid Cells/immunology , Toll-Like Receptor 4/deficiency , p38 Mitogen-Activated Protein Kinases/metabolism
13.
J Vasc Surg ; 58(2): 460-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23414695

ABSTRACT

BACKGROUND: We have previously shown that the danger signal high-mobility group box 1 (HMGB1) promotes angiogenesis when administered to ischemic muscle. HMGB1 signals through Toll-like receptor 4 (TLR4) as well as the receptor for advanced glycation end-products (RAGE). However, the actions of these receptors in ischemic injury and muscle recovery are not known. We hypothesize that TLR4 mediates tissue recovery and angiogenesis in response to ischemia. METHODS: Femoral artery ligation was performed in control, TLR4 competent (C3H/HeOuJ) and incompetent (C3H/HeJ) mice, as well as RAGE knockout mice and their C57B6 control counterparts. In other experiments, control mice were pretreated with anti-HMGB1 neutralizing antibody before femoral artery ligation. After 2 weeks, limb perfusion was evaluated using laser Doppler perfusion imaging and reported as the ratio of blood flow in the ischemic to nonischemic limb. Muscle necrosis, fat replacement, and vascular density in the anterior tibialis muscle were quantified histologically. In vitro, TLR4 and RAGE expression was evaluated in human dermal microvascular endothelial cells in response to hypoxia. Human dermal microvascular endothelial cells treated with HMGB1 alone and in the presence of anti-TLR4 antibody were probed for phosphorylated extracellular signal-regulated kinase (ERK), a signaling molecule critical to endothelial cell (EC) angiogenic behavior. RESULTS: Both anti-HMGB1 antibody as well as defective TLR4 signaling in HeJ mice resulted in prominent muscle necrosis 2 weeks after femoral artery ligation. Control HeOuJ mice had less necrosis than TLR4 incompetent HeJ mice, but a greater amount of fat replacement. In contrast to control C3H mice, control C57B6 mice demonstrated prominent muscle regeneration with very little necrosis. Muscle regeneration was not dependent on RAGE. While vascular density did not differ between strains, mice with intact RAGE and TLR4 signaling had less blood flow in ischemic limbs compared with mutant strains. In vitro, EC TLR4 expression increased in response to hypoxia while TLR4 antagonism decreased HMGB1-induced activation of extracellular signal-regulated kinase. CONCLUSIONS: Both HMGB1 and TLR4 protect against muscle necrosis after hindlimb ischemia. However, muscle regeneration does not appear to be tied to vascular density. HMGB1 likely activates angiogenic behavior in ECs in vitro, and this activation may be modulated by TLR4. The improvement in blood flow seen in mice with absent TLR4 and RAGE signaling may suggest anti-angiogenic roles for both receptors, or vasoconstriction induced by TLR4 and RAGE mediated inflammatory pathways.


Subject(s)
HMGB1 Protein/metabolism , Ischemia/metabolism , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Neovascularization, Physiologic , Toll-Like Receptor 4/metabolism , Animals , Antibodies, Neutralizing/administration & dosage , Cells, Cultured , Disease Models, Animal , Endothelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Femoral Artery/surgery , HMGB1 Protein/immunology , Hindlimb , Humans , Ischemia/genetics , Ischemia/pathology , Ischemia/physiopathology , Laser-Doppler Flowmetry , Ligation , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , Necrosis , Perfusion Imaging/methods , Phosphorylation , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Regeneration , Regional Blood Flow , Signal Transduction , Time Factors , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
14.
Mycopathologia ; 174(1): 21-30, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22286932

ABSTRACT

For many fungal diseases, macrophages are the major cell population implicated in host protection, primarily by their ability to eliminate the invading fungal pathogen through phagocytosis. In sporotrichosis, this remains true, because of macrophages' ability to recognize Sporothrix schenckii through specific receptors for some of the fungus' cellular surface constituents. Further confirmation for macrophages' pivotal role in fungal diseases came with the identification of toll-like receptors, and the subsequent numerous associations found between TLR-4 deficiency and host susceptibility to diverse fungal pathogens. Involvement of TLR-4 in immune response against sporotrichosis has been conducted to investigate how TLR-4 signaling could affect inflammatory response development through evaluation of H2O2 production and IL-1ß, IL-6 and TGF-ß release during the course of S. schenckii infection on TLR-4-deficient mice. The results showed that macrophages are largely dependent on TLR-4 for inflammatory activation and that in the absence of TLR-4 signaling, increased TGF-ß release may be one of the contributing factors for the abrogated inflammatory activation of peritoneal exudate cells during mice sporotrichosis.


Subject(s)
Macrophages/immunology , Macrophages/microbiology , Sporothrix/immunology , Sporothrix/pathogenicity , Sporotrichosis/immunology , Sporotrichosis/pathology , Toll-Like Receptor 4/deficiency , Animals , Antifungal Agents/metabolism , Antifungal Agents/toxicity , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/toxicity , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Mice , Mice, Knockout , Transforming Growth Factor beta/metabolism
15.
J Leukoc Biol ; 91(2): 285-98, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22075930

ABSTRACT

Osteoarticular complications are common in human brucellosis, but the pathogenic mechanisms involved are largely unknown. In this manuscript, we described an immune mechanism for inflammatory bone loss in response to infection by Brucella abortus. We established a requirement for MyD88 and TLR2 in TNF-α-elicited osteoclastogenesis in response to B. abortus infection. CS from macrophages infected with B. abortus induced BMM to undergo osteoclastogenesis. Although B. abortus-infected macrophages actively secreted IL-1ß, IL-6, and TNF-α, osteoclastogenesis depended on TNF-α, as CS from B. abortus-infected macrophages failed to induce osteoclastogenesis in BMM from TNFRp55⁻/⁻ mice. CS from B. abortus-stimulated MyD88⁻/⁻ and TLR2⁻/⁻ macrophages failed to express TNF-α, and these CS induced no osteoclast formation compared with that of the WT or TLR4⁻/⁻ macrophages. Omp19, a B. abortus lipoprotein model, recapitulated the cytokine production and subsequent osteoclastogenesis induced by the whole bacterium. All phenomena were corroborated using human monocytes, indicating that this mechanism could play a role in human osteoarticular brucellosis. Our results indicate that B. abortus, through its lipoproteins, may be involved in bone resorption through the pathological induction of osteoclastogenesis.


Subject(s)
Brucella abortus/physiology , Macrophages, Peritoneal/physiology , Myeloid Differentiation Factor 88/physiology , Osteoclasts/physiology , Toll-Like Receptor 2/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Antigens, Bacterial/pharmacology , Bacterial Outer Membrane Proteins/pharmacology , Brucellosis/immunology , Cell Differentiation/drug effects , Cells, Cultured/drug effects , Cells, Cultured/physiology , Culture Media, Conditioned/pharmacology , Cytokines/biosynthesis , Female , Humans , Lipoproteins/pharmacology , Macrophages, Peritoneal/drug effects , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/physiology , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , RANK Ligand/biosynthesis , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/deficiency , Transforming Growth Factor beta/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis
16.
J Immunol ; 186(1): 411-22, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21098233

ABSTRACT

Leishmania major is a protozoan parasite that causes skin ulcerations in cutaneous leishmaniasis. In the mammalian host, the parasite resides in professional phagocytes and has evolved to avoid killing by macrophages. We identified L. major genes encoding inhibitors of serine peptidases (ISPs), which are orthologs of bacterial ecotins, and found that ISP2 inhibits trypsin-fold S1A family peptidases. In this study, we show that L. major mutants deficient in ISP2 and ISP3 (Δisp2/3) trigger higher phagocytosis by macrophages through a combined action of the complement type 3 receptor, TLR4, and unregulated activity of neutrophil elastase (NE), leading to parasite killing. Whereas all three components are required to mediate enhanced parasite uptake, only TLR4 and NE are necessary to promote parasite killing postinfection. We found that the production of superoxide by macrophages in the absence of ISP2 is the main mechanism controlling the intracellular infection. Furthermore, we show that NE modulates macrophage infection in vivo, and that the lack of ISP leads to reduced parasite burdens at later stages of the infection. Our findings support the hypothesis that ISPs function to prevent the activation of TLR4 by NE during the Leishmania-macrophage interaction to promote parasite survival and growth.


Subject(s)
Intracellular Fluid/parasitology , Leishmania major/enzymology , Leishmania major/growth & development , Leukocyte Elastase/physiology , Macrophages, Peritoneal/parasitology , Serine Proteinase Inhibitors/physiology , Serpins/physiology , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/metabolism , Animals , Cells, Cultured , Host-Parasite Interactions/immunology , Intracellular Fluid/enzymology , Intracellular Fluid/immunology , Leishmania major/immunology , Leukocyte Elastase/antagonists & inhibitors , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/immunology , Serine Proteinase Inhibitors/deficiency , Serine Proteinase Inhibitors/genetics , Serpins/deficiency , Serpins/genetics , Toll-Like Receptor 4/deficiency
17.
J Neuroimmunol ; 225(1-2): 62-7, 2010 Aug 25.
Article in English | MEDLINE | ID: mdl-20554031

ABSTRACT

Systemic low doses of the endotoxin lipopolysaccharide (LPS) administered at CT15 (circadian time 12 corresponds to locomotor activity onset) induce phase delays of locomotor activity rhythms in mice. To evaluate if this effect was mediated by the Toll-like receptor 4 (TLR4), our present aim was to characterize the circadian behavior and LPS-induced circadian response of TLR4 (LPS receptor)-deficient mice (in C57bl/10 and C3H backgrounds). In mutants, we observed a free-running period and a light-induced phase delay similar to the one observed in their corresponding wild-type (WT) littermates. The LPS-induced phase delay, wheel running inhibition and c-Fos/Per-1 immunoreactivity in the paraventricular nuclei observed in WT mice was absent or significantly decreased in the TLR4-deficient mice. In conclusion, we show that LPS-induced circadian responses are mediated by TLR4.


Subject(s)
Circadian Rhythm/drug effects , Circadian Rhythm/genetics , Lipopolysaccharides/pharmacology , Motor Activity/drug effects , Motor Activity/genetics , Toll-Like Receptor 4/physiology , Analysis of Variance , Animals , Behavior, Animal/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Species Specificity , Suprachiasmatic Nucleus/drug effects , Suprachiasmatic Nucleus/metabolism , Toll-Like Receptor 4/deficiency
18.
PLoS Pathog ; 6(4): e1000870, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20442858

ABSTRACT

The murine model of T. cruzi infection has provided compelling evidence that development of host resistance against intracellular protozoans critically depends on the activation of members of the Toll-like receptor (TLR) family via the MyD88 adaptor molecule. However, the possibility that TLR/MyD88 signaling pathways also control the induction of immunoprotective CD8+ T cell-mediated effector functions has not been investigated to date. We addressed this question by measuring the frequencies of IFN-gamma secreting CD8+ T cells specific for H-2K(b)-restricted immunodominant peptides as well as the in vivo Ag-specific cytotoxic response in infected animals that are deficient either in TLR2, TLR4, TLR9 or MyD88 signaling pathways. Strikingly, we found that T. cruzi-infected Tlr2(-/-), Tlr4(-/-), Tlr9(-/) (-) or Myd88(-/-) mice generated both specific cytotoxic responses and IFN-gamma secreting CD8+ T cells at levels comparable to WT mice, although the frequency of IFN-gamma+CD4+ cells was diminished in infected Myd88(-/-) mice. We also analyzed the efficiency of TLR4-driven immune responses against T. cruzi using TLR4-deficient mice on the C57BL genetic background (B6 and B10). Our studies demonstrated that TLR4 signaling is required for optimal production of IFN-gamma, TNF-alpha and nitric oxide (NO) in the spleen of infected animals and, as a consequence, Tlr4(-/-) mice display higher parasitemia levels. Collectively, our results indicate that TLR4, as well as previously shown for TLR2, TLR9 and MyD88, contributes to the innate immune response and, consequently, resistance in the acute phase of infection, although each of these pathways is not individually essential for the generation of class I-restricted responses against T. cruzi.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chagas Disease/immunology , Immunity, Innate , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Animals , Chagas Disease/genetics , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Nitric Oxide Synthase Type II/immunology , Nitric Oxide Synthase Type II/metabolism , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Trypanosoma cruzi/immunology
19.
Med Mycol ; 48(8): 1009-17, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20465519

ABSTRACT

Candida albicans is recognized by phagocytic cells through a set of recognition receptors patterns. Recently, we showed the importance of TLR2 in the regulation of neutrophil survival after C. albicans infection. In the present work, we analyzed the involvement of TLR4 in the recognition of C. albicans by neutrophils and macrophages. Our results show that the absence of functional TLR4 resulted in lower chemotaxis of neutrophils to the site of infection, lower levels of TNF-α, CXCL1 and nitric oxide, and dissemination and persistence of the pathogen in lymph nodes and spleen. In vitro, the phagocytic activity, nitric oxide production and myeloperoxidase activity, CXCL1, IL-1ß production by neutrophils from TLR4-defective mice were not changed. In contrast, macrophages from TLR4-defective mice demonstrated lower phagocytosis and lower levels of CXCL1, IL-1ß and TNF-α. Together, these data demonstrate that TLR4 signals are important for the recognition of C. albicans by macrophages and their absence allows persistence of the infection.


Subject(s)
Candida albicans/immunology , Candidiasis/immunology , Macrophages/immunology , Toll-Like Receptor 4/immunology , Animals , Cell Movement , Chemokine CXCL1/metabolism , Chemotaxis , Disease Models, Animal , Interleukin-1beta/metabolism , Lymph Nodes/microbiology , Macrophages/microbiology , Male , Mice , Mice, Inbred C3H , Mice, Knockout , Neutrophils/immunology , Neutrophils/microbiology , Nitric Oxide/metabolism , Spleen/microbiology , Toll-Like Receptor 4/deficiency , Tumor Necrosis Factor-alpha/metabolism
20.
Shock ; 34(3): 306-13, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20160673

ABSTRACT

Intestinal ischemia-reperfusion (I/R) injury may cause acute systemic and lung inflammation. Here, we revisited the role of TNF-alpha in an intestinal I/R model in mice, showing that this cytokine is not required for the local and remote inflammatory response upon intestinal I/R injury using neutralizing TNF-alpha antibodies and TNF ligand-deficient mice. We demonstrate increased neutrophil recruitment in the lung as assessed by myeloperoxidase activity and augmented IL-6, granulocyte colony-stimulating factor, and KC levels, whereas TNF-alpha levels in serum were not increased and only minimally elevated in intestine and lung upon intestinal I/R injury. Importantly, TNF-alpha antibody neutralization neither diminished neutrophil recruitment nor any of the cytokines and chemokines evaluated. In addition, the inflammatory response was not abrogated in TNF and TNF receptors 1 and 2-deficient mice. However, in view of the damage on the intestinal barrier upon intestinal I/R with systemic bacterial translocation, we asked whether Toll-like receptor (TLR) activation is driving the inflammatory response. In fact, the inflammatory lung response is dramatically reduced in TLR2/4-deficient mice, confirming an important role of TLR receptor signaling causing the inflammatory lung response. In conclusion, endogenous TNF-alpha is not or minimally elevated and plays no role as a mediator for the inflammatory response upon ischemic tissue injury. By contrast, TLR2/4 signaling induces an orchestrated cytokine/chemokine response leading to local and remote pulmonary inflammation, and therefore disruption of TLR signaling may represent an alternative therapeutic target.


Subject(s)
Intestines/blood supply , Pneumonia/physiopathology , Reperfusion Injury/complications , Respiratory Distress Syndrome/physiopathology , Toll-Like Receptor 2/physiology , Toll-Like Receptor 4/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Antibodies, Monoclonal/pharmacology , Chemotaxis, Leukocyte , Cytokines/blood , Intestines/enzymology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/enzymology , Peroxidase/analysis , Pneumonia/etiology , Reperfusion Injury/physiopathology , Respiratory Distress Syndrome/etiology , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL