Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Mol Neurosci ; 70(1): 71-83, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31478134

ABSTRACT

The disruption of the blood-brain barrier (BBB) and the consequent brain edema are major contributors to the pathogenesis of cerebral ischemia/reperfusion injury. RhoA is generally thought to play a crucial role in the process of BBB disruption and participate in the signaling pathways emanating from TLR4. However, it remains unverified the regulatory role of TLR4 in the RhoA/ROCK pathway in cerebral I/R injury and its effects on the BBB as well. The present study probes into the protective effect of ANF on the BBB after cerebral I/R injury and the possible mechanisms. Focal cerebral ischemia was induced by 120 min of transient middle cerebral artery occlusion (MCAO). ANF (1, 2, 4 µg/kg) was achieved by intravenous injection after 120 min of MCAO followed by 1, 24, 48, and 72 h reperfusion. Evans blue extravasation, brain water content, RhoA activity, and the expressions of TLR4, ROCK1/2, p-MLC2, MMP-2/9, ZO-1, occludin, and claudin-5 protein in rat brain were evaluated 72 h after reperfusion. ANF could significantly reduce the Evans blue extravasation and water content in the ipsilateral hemisphere and obviously increase the occludin, claudin-5, and ZO-1 expression after cerebral I/R injury. Furthermore, cerebral I/R injury induced apparently increased expression of TLR4, RhoA-GTP, ROCK1/2, p-MLC2, and MMMP-2/9, which, however, could be remarkably alleviated by ANF intervention. Taken together, the TLR4/RhoA/ROCK signaling pathway is implicated in BBB breakdown after cerebral I/R injury, and ANF preserves BBB integrity, probably via inhibiting the TLR4/RhoA/ROCK signaling pathway.


Subject(s)
Blood-Brain Barrier/drug effects , Crotalid Venoms/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Lectins, C-Type/therapeutic use , Neuroprotective Agents/therapeutic use , Reperfusion Injury/drug therapy , Toll-Like Receptor 4/metabolism , Animals , Blood-Brain Barrier/metabolism , Cardiac Myosins/metabolism , Crotalid Venoms/administration & dosage , Crotalid Venoms/pharmacology , Lectins, C-Type/administration & dosage , Male , Matrix Metalloproteinases/metabolism , Myosin Light Chains/metabolism , Neuroprotective Agents/adverse effects , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism
2.
J Virol ; 93(19)2019 10 01.
Article in English | MEDLINE | ID: mdl-31315995

ABSTRACT

CD69 is highly expressed on the leukocyte surface upon viral infection, and its regulatory role in the vaccinia virus (VACV) immune response has been recently demonstrated using CD69-/- mice. Here, we show augmented control of VACV infection using the anti-human CD69 monoclonal antibody (MAb) 2.8 as both preventive and therapeutic treatment for mice expressing human CD69. This control was related to increased natural killer (NK) cell reactivity and increased numbers of cytokine-producing T and NK cells in the periphery. Moreover, similarly increased immunity and protection against VACV were reproduced over both long and short periods in anti-mouse CD69 MAb 2.2-treated immunocompetent wild-type (WT) mice and immunodeficient Rag2-/- CD69+/+ mice. This result was not due to synergy between infection and anti-CD69 treatment since, in the absence of infection, anti-human CD69 targeting induced immune activation, which was characterized by mobilization, proliferation, and enhanced survival of immune cells as well as marked production of several innate proinflammatory cytokines by immune cells. Additionally, we showed that the rapid leukocyte effect induced by anti-CD69 MAb treatment was dependent on mTOR signaling. These properties suggest the potential of CD69-targeted therapy as an antiviral adjuvant to prevent derived infections.IMPORTANCE In this study, we demonstrate the influence of human and mouse anti-CD69 therapies on the immune response to VACV infection. We report that targeting CD69 increases the leukocyte numbers in the secondary lymphoid organs during infection and improves the capacity to clear the viral infection. Targeting CD69 increases the numbers of gamma interferon (IFN-γ)- and tumor necrosis factor alpha (TNF-α)-producing NK and T cells. In mice expressing human CD69, treatment with an anti-CD69 MAb produces increases in cytokine production, survival, and proliferation mediated in part by mTOR signaling. These results, together with the fact that we have mainly worked with a human-CD69 transgenic model, reveal CD69 as a treatment target to enhance vaccine protectiveness.


Subject(s)
Immunologic Factors/antagonists & inhibitors , Killer Cells, Natural/immunology , Lectins, C-Type/antagonists & inhibitors , T-Lymphocytes/immunology , Vaccinia virus/immunology , Vaccinia/prevention & control , Animals , Antibodies, Monoclonal/administration & dosage , Antigens, CD/administration & dosage , Antigens, CD/genetics , Antigens, Differentiation, T-Lymphocyte/administration & dosage , Antigens, Differentiation, T-Lymphocyte/genetics , Disease Models, Animal , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/genetics , Lectins, C-Type/administration & dosage , Lectins, C-Type/genetics , Mice , Mice, Transgenic , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Vaccinia/immunology , Vaccinia/therapy
3.
J Anat ; 230(1): 30-46, 2017 01.
Article in English | MEDLINE | ID: mdl-27757964

ABSTRACT

Multiple sclerosis (MS) is a chronic neurological disorder that affects the central nervous system (CNS), and results in CNS inflammation and damage to myelin. In this study, we examined the possible synergistic effects of C16, angiopoietin-1 (Ang-1) and regeneration gene protein 2 (Reg-2) in alleviating inflammation in an acute experimental autoimmune encephalomyelitis (EAE) model. We employed multiple histological, morphological and iconographic assays to examine the effect of those drugs on disease onset, clinical scores and behavioral deficits. Our results demonstrated that triple combination therapy was more efficient than the monotherapy in EAE treatment. The triple therapy significantly delayed the onset of motor symptoms, reduced disease severity, attenuated inflammatory cell infiltration and suppressed the secretion of proinflammatory cytokines. Additionally, treatment increased anti-inflammatory cytokines expression, inhibited reactive astrocytes proliferation, reduced demyelination and axonal loss, and finally reduced the neural death. Specifically, Reg-2 administration rescued oligodendrocytes and neuronal axons mainly by direct neurotrophic effects, while C16+Ang-1 (C+A) mainly improved the inflammatory milieu. In conclusion, our study suggests a possible synergistic effect through targeting a variety of pathways in relieving the clinical symptoms of inflammation in acute EAE model. Therefore, using molecules that target different molecular pathways can be beneficial for exploring novel therapeutic approaches for MS treatment.


Subject(s)
Angiopoietin-1/administration & dosage , Antigens, Neoplasm/administration & dosage , Biomarkers, Tumor/administration & dosage , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Lectins, C-Type/administration & dosage , Peptide Fragments/administration & dosage , Animals , Disease Models, Animal , Drug Therapy, Combination , Inflammation/pathology , Inflammation/prevention & control , Male , Pancreatitis-Associated Proteins , Peptide Fragments/genetics , Rats , Rats, Inbred Lew
4.
Toxicol Sci ; 150(2): 378-89, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26781512

ABSTRACT

High density lipoprotein (HDL)-targeted therapies, which promote cholesterol efflux from cells, are currently in development for reducing cardiovascular events in acute coronary syndrome. Human apolipoprotein A-I (apoA-I), the major HDL protein, was fused to the trimerization domain of tetranectin (TN) and complexed with phospholipids to generate a HDL mimetic (lipidated TN-ApoA-I) with reduced renal clearance and enhanced efficacy. Cynomolgus monkeys received 24-h intravenous infusions of control, 100 mg/kg or 400 mg/kg lipidated TN-ApoA-I every 4 days for 3 weeks, followed by a 6-week recovery period. After multiple infusions of lipidated TN-ApoA-I, clinical condition deteriorated and was accompanied by changes indicative of a progressive inflammatory response; increased levels of cytokines, C-reactive protein and vascular/perivascular infiltrates in multiple tissues. Rapid formation of antidrug antibodies occurred in all animals receiving lipidated TN-ApoA-I. Enhanced drug clearance corresponding to a relative lack of high molecular weight immune complexes in blood, suggestive of preferred removal/clearance, was observed in some animals. Expected dose-dependent increases in serum lipids were accompanied by vacuolated monocytes/macrophages in multiple organs, which in the glomeruli were shown to be CD68-positive, contain lipid and co-localized with granular IgG deposits. Lipid accumulation may have been a direct result of a high drug load, possibly enhanced by immune complex formation, inflammation, and altered lipid metabolism. Noteworthy was the inter- individual inconsistency in the severity of clinical and histopathologic findings, drug clearance and inflammatory markers. In conclusion, multiple infusions of lipidated TN-ApoA-I resulted in high immunogenicity, lipid accumulation and were not well tolerated in nonhuman primates.


Subject(s)
Antigen-Antibody Complex/blood , Apolipoprotein A-I/toxicity , Lectins, C-Type/administration & dosage , Lipids/blood , Recombinant Fusion Proteins/toxicity , Animals , Apolipoprotein A-I/administration & dosage , Apolipoprotein A-I/immunology , Apolipoprotein A-I/pharmacokinetics , C-Reactive Protein/analysis , Cytokines/blood , Dose-Response Relationship, Drug , Fibrin Fibrinogen Degradation Products/analysis , Fibrinogen/analysis , Inflammation/blood , Inflammation/chemically induced , Infusions, Intravenous , Lectins, C-Type/immunology , Lipids/immunology , Macaca fascicularis , Male , Metabolic Clearance Rate , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacokinetics
5.
Mol Cell Endocrinol ; 422: 150-159, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26683606

ABSTRACT

Regenerating gene 3α (Reg3α) protein is a trophic factor that stimulates cell and tissue proliferation, neogenesis and also acts against apoptosis and necrosis. In order to explore the potential roles of recombinant Reg3α (rReg3α), we produced a mature rReg3α polypeptide for direct administration in l-arginine (L-Arg) induced acute pancreatitis (AP) in mice. Our results showed that rReg3α stimulated cell proliferation through Erk1/2 and p38 phosphorylation and also cyclin D1 upregulation mediated by Akt/ATF-2 signaling. Moreover, rReg3α administration significantly reduced the pancreatic damage caused by L-Arg injection, as shown in histological examination and serum amylase, lipase and C-reactive protein (CRP) assays. Not only acinar cell necrosis but also apoptosis found in the pancreas of AP mice were alleviated by rReg3α. Finally, upregulated Bcl-2 and Bcl-xL and suppressed poly (ADP-ribose) synthetase/polymerase (PARP) levels were detected as being relevant to the mechanism of rReg3α protection. We therefore conclude that rReg3α acts as a protective polypeptide against AP in mice by enhancing Bcl-2 and Bcl-xL expressions and suppressing PARP level.


Subject(s)
Antigens, Neoplasm/administration & dosage , Arginine/adverse effects , Biomarkers, Tumor/administration & dosage , Lectins, C-Type/administration & dosage , Pancreatitis/prevention & control , Proto-Oncogene Proteins c-bcl-2/metabolism , Acinar Cells/drug effects , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Antigens, Neoplasm/pharmacology , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/pharmacology , Cell Line , Cell Proliferation , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Mice , Pancreatitis/chemically induced , Pancreatitis/pathology , Pancreatitis-Associated Proteins , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Signal Transduction/drug effects
6.
Eur J Immunol ; 46(3): 689-700, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26678013

ABSTRACT

The development of new immunization strategies requires a better understanding of early molecular and cellular events occurring at the site of injection. The skin is particularly rich in immune cells and represents an attractive site for vaccine administration. Here, we specifically targeted vaccine antigens to epidermal Langerhans cells (LCs) using a fusion protein composed of HIV antigens and a monoclonal antibody targeting Langerin. We developed a fluorescence imaging approach to visualize, in vivo, the vaccine-targeted cells. Studies were performed in nonhuman primates (NHPs) because of their relevance as a model to assess human vaccines. We directly demonstrated that in NHPs, intradermally injected anti-Langerin-HIVGag specifically targets epidermal LCs and induces rapid changes in the LC network, including LC activation and migration out of the epidermis. Vaccine targeting of LCs significantly improved anti-HIV immune response without requirement of an adjuvant. Although the co-injection of the TLR-7/8 synthetic ligand, R-848 (resiquimod), with the vaccine, did not enhance significantly the antibody response, it stimulated recruitment of HLA-DR+ inflammatory cells to the site of immunization. This study allowed us to characterize the dynamics of early local events following the injection of a vaccine-targeted epidermal LCs and R-848.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antigens, CD/immunology , Langerhans Cells/immunology , Lectins, C-Type/immunology , Mannose-Binding Lectins/immunology , Vaccines/administration & dosage , Animals , Antibodies, Viral/blood , Antigens, CD/administration & dosage , Epidermal Cells , Epidermis/immunology , HIV Core Protein p24/administration & dosage , HIV Core Protein p24/immunology , Humans , Imidazoles/administration & dosage , Imidazoles/immunology , Injections, Intradermal , Intravital Microscopy , Langerhans Cells/ultrastructure , Lectins, C-Type/administration & dosage , Macaca fascicularis , Mannose-Binding Lectins/administration & dosage , Optical Imaging , Vaccines/immunology
7.
Thromb Res ; 134(5): 1150-6, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25281435

ABSTRACT

This is the first report on the characterization of a snaclec (RVsnaclec) purified from Daboia russelii russelii venom. The RVsnaclec is a heterodimer of two subunits, α (15.1 kDa) and ß (9 kDa). These subunits are covalently linked to form multimeric (αß)2 and (αß)4 structures. Peptide mass fingerprinting analysis of RVsnaclec via LC-MS/MS demonstrated its similarity to snaclecs purified from other viperid snake venoms. Two tryptic peptide sequences of RVsnaclec revealed the putative conserved domains of C-type lectin (CTL). RVsnaclec dose-dependently increased the Ca-clotting time and prothrombin time of platelet-poor plasma (PPP); however, it did not affect the partial thromboplastin time (APTT) or thrombin time of PPP. The in vitro and in vivo anticoagulant activity of RVsnaclec is correlated to its binding and subsequent uncompetitive inhibition of FXa (Ki = 0.52 µmole) in a Ca(2+)-independent manner; however, supplementation with 0.25 mM Ca(2+) enhanced the Xa binding potency of RVsnaclec. Monovalent or polyvalent antivenom failed to neutralize its anticoagulant potency, and RVsnaclec did not inhibit trypsin, chymotrypsin, thrombin or plasmin. RVsnaclec was devoid of hemolytic activity or cytotoxicity against several human cancer cell lines, demonstrated concentration-dependent aggregation and deaggregation of human platelets, and inhibited the ADP-induced aggregation of platelet. RVsnaclec (5.0 mg/kg body weight) was non-lethal to mice and showed no adverse pharmacological effects, suggesting that it has potential as a lead compound for future therapeutic applications in cardiovascular disorders.


Subject(s)
Anticoagulants/pharmacology , Daboia , Lectins, C-Type/chemistry , Viper Venoms/pharmacology , Animals , Anticoagulants/administration & dosage , Anticoagulants/chemistry , Anticoagulants/toxicity , Blood Coagulation/drug effects , Blood Platelets/cytology , Blood Platelets/drug effects , Blood Platelets/metabolism , Calcium/metabolism , Factor Xa/metabolism , Factor Xa Inhibitors/administration & dosage , Factor Xa Inhibitors/chemistry , Factor Xa Inhibitors/pharmacology , Factor Xa Inhibitors/toxicity , Goats , Humans , Lectins, C-Type/administration & dosage , Lectins, C-Type/isolation & purification , Mice , Platelet Aggregation/drug effects , Daboia/metabolism , Viper Venoms/administration & dosage , Viper Venoms/chemistry , Viper Venoms/toxicity
8.
Fish Shellfish Immunol ; 38(2): 294-302, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24721287

ABSTRACT

This study investigated the use of a recombinant protein of Neoparamoeba perurans, the causative agent of Amoebic gill disease (AGD), as an immunogen to generate systemic and mucosal antibody responses against the parasite. Genes encoding N. perurans homologs of mannose-binding protein (MBP) from Acanthamoeba spp. have been identified. From these, a Neoparamoeba MBP - like EST has been identified and produced as a recombinant fusion protein. Attachment of N. perurans to the gill might be reduced by antibody-mediated interference of this protein, but this is dependent on the presence and level of functional antibodies in the mucus. Fish were immunized with the protein via i.p. injection with Freund's complete adjuvant (FCA); and serum and skin mucus samples were collected before and after immunization. Antibodies (IgM) present in samples were characterized via Western blot and their levels measured with an ELISA. The immunization was able to induce a systemic IgM response 8 weeks after primary exposure and a mucosal response 4 weeks post initial immunization, which were specific to the recombinant protein but not to antigens obtained from crude amoebic preparations. However, adherence of the antibodies to the parasite was observed using immunocytochemistry, and both, serum and skin mucus IgM, were able to bind the surface of formalin-fixed N. perurans. This finding may contribute to further research into the development of a vaccine for AGD.


Subject(s)
Amebiasis/veterinary , Amoebozoa/drug effects , Fish Diseases/therapy , Immunity, Humoral/drug effects , Protozoan Proteins/immunology , Recombinant Proteins/pharmacology , Salmo salar , Amebiasis/parasitology , Amebiasis/therapy , Amino Acid Sequence , Animals , Antibodies, Protozoan/metabolism , Fish Diseases/parasitology , Lectins, C-Type/administration & dosage , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Microscopy, Fluorescence/veterinary , Protozoan Proteins/administration & dosage , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/metabolism
9.
J Immunol ; 190(8): 4116-28, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23514738

ABSTRACT

In a previous attempt to generate a protective vaccine against Candida albicans, a ß-mannan tetanus toxoid conjugate showed poor immunogenicity in mice. To improve the specific activation toward the fungal pathogen, we aimed to target Dectin-1, a pattern-recognition receptor expressed on monocytes, macrophages, and dendritic cells. Laminarin, a ß-glucan ligand of Dectin-1, was incorporated into the original ß-mannan tetanus toxoid conjugate providing a tricomponent conjugate vaccine. A macrophage cell line expressing Dectin-1 was employed to show binding and activation of Dectin-1 signal transduction pathway by the ß-glucan-containing vaccine. Ligand binding to Dectin-1 resulted in the following: 1) activation of Src family kinases and Syk revealed by their recruitment and phosphorylation in the vicinity of bound conjugate and 2) translocation of NF-κB to the nucleus. Treatment of immature bone marrow-derived dendritic cells (BMDCs) with tricomponent or control vaccine confirmed that the ß-glucan-containing vaccine exerted its enhanced activity by virtue of dendritic cell targeting and uptake. Immature primary cells stimulated by the tricomponent vaccine, but not the ß-mannan tetanus toxoid vaccine, showed activation of BMDCs. Moreover, treated BMDCs secreted increased levels of several cytokines, including TGF-ß and IL-6, which are known activators of Th17 cells. Immunization of mice with the novel type of vaccine resulted in improved immune response manifested by high titers of Ab recognizing C. albicans ß-mannan Ag. Vaccine containing laminarin also affected distribution of IgG subclasses, showing that vaccine targeting to Dectin-1 receptor can benefit from augmentation and immunomodulation of the immune response.


Subject(s)
Dendritic Cells/metabolism , Drug Delivery Systems , Lectins, C-Type/administration & dosage , Tetanus Toxoid/administration & dosage , Tetanus Toxoid/immunology , beta-Glucans/metabolism , Animals , Binding Sites/immunology , Cell Line , Dendritic Cells/immunology , Drug Delivery Systems/methods , Epitopes/immunology , Epitopes/metabolism , Glucans , Lectins, C-Type/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Polysaccharides/immunology , Polysaccharides/metabolism , Tetanus Toxoid/metabolism , Trisaccharides/administration & dosage , Trisaccharides/immunology , Trisaccharides/metabolism , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/immunology , Vaccines, Conjugate/metabolism , beta-Glucans/immunology
10.
Nat Chem Biol ; 9(4): 250-6, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23416331

ABSTRACT

Targeted delivery of antigens to dendritic cells (DCs) is a promising vaccination strategy. However, to ensure immunity, the approach depends on coadministration of an adjuvant. Here we ask whether targeting of both adjuvant and antigen to DCs is sufficient to induce immunity. Using a protein ligation method, we develop a general approach for linking the immune stimulant, poly dA:dT (pdA:dT), to a monoclonal antibody (mAb) specific for DEC205 (DEC). We show that DEC-specific mAbs deliver pdA:dT to DCs for the efficient production of type I interferon in human monocyte-derived DCs and in mice. Notably, adaptive T-cell immunity is elicited when mAbs specific for DEC-pdA:dT are used as the activation stimuli and are administered together with a DC-targeted antigen. Collectively, our studies indicate that DCs can integrate innate and adaptive immunity in vivo and suggest that dual delivery of antigen and adjuvant to DCs might be an efficient approach to vaccine development.


Subject(s)
Adaptive Immunity/drug effects , Antibodies, Monoclonal/immunology , Antigens, CD/immunology , Antigens/immunology , Dendritic Cells/drug effects , Immunity, Innate/drug effects , Immunoconjugates/immunology , Lectins, C-Type/immunology , Poly dA-dT/immunology , Receptors, Cell Surface/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Antigens/administration & dosage , Antigens/chemistry , Antigens, CD/administration & dosage , Antigens, CD/chemistry , Dendritic Cells/immunology , Drug Delivery Systems , Genetic Vectors , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/chemistry , Interferon Type I/biosynthesis , Interferon Type I/immunology , Lectins, C-Type/administration & dosage , Lectins, C-Type/chemistry , Mice , Mice, Inbred C57BL , Minor Histocompatibility Antigens , Plasmids , Poly dA-dT/administration & dosage , Poly dA-dT/chemistry , Receptors, Cell Surface/administration & dosage , Receptors, Cell Surface/chemistry
11.
Toxicon ; 63: 19-31, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23159397

ABSTRACT

Snakebite envenoming by Bothrops caribbaeus, an endemic viperid from the Lesser Antillean island of Saint Lucia, is clinically characterized by local tissue damage and systemic thrombosis that can lead to cerebral, myocardial or pulmonary infarctions and venous thromboses. Systemic effects (lethality, pulmonary hemorrhage, thrombocytopenia and coagulopathy) induced by intravenous (i.v.) administration of B. caribbaeus venom were studied in mice. The role of snake venom metalloproteinases (SVMPs) in these systemic alterations was assessed by inhibition with the chelating agent calcium disodium ethylenediaminetetraacetic acid (CaNa(2)EDTA). A snake C-type lectin-like (snaclec) and a type P-III hemorrhagic SVMP were isolated and characterized from this venom, and the effect of venom and the isolated snaclec on human platelet aggregation was studied in vitro. Results indicate that SVMPs play an important role in the overall toxicity of B. caribbaeus venom, being responsible for systemic hemorrhage and lethality, but not thrombocytopenia, whereas the isolated snaclec is involved in the thrombocytopenic effect. Both venom and snaclec induce platelet aggregation/agglutination. Moreover, the snaclec binds directly to glycoprotein Ib (GPIb) and induces agglutination in washed fixed platelets. On the other hand, B. caribbaeus venom hydrolyzed fibrinogen in vitro and induced a partial drop of fibrinogen levels with an increase in fibrin/fibrinogen degradation products (FDP) levels in vivo. The negative result for D-dimer (DD) in plasma is consistent with the lack of microscopic evidence of pulmonary thrombosis and endothelial cell damage. Likewise, no increments in plasma sE-selectin levels were detected. The absence of thrombosis in this murine model suggests that this effect may be species-specific.


Subject(s)
Bothrops/physiology , Coagulants/toxicity , Crotalid Venoms/toxicity , Thrombocytopenia/chemically induced , Thrombosis/chemically induced , Animals , Coagulants/chemistry , Crotalid Venoms/chemistry , Crotalid Venoms/enzymology , Disease Models, Animal , Edetic Acid/chemistry , Fibrinogen/drug effects , Hemorrhage/chemically induced , Hemorrhage/pathology , Humans , Injections, Intravenous , Lectins, C-Type/administration & dosage , Lectins, C-Type/chemistry , Longevity/drug effects , Lung Diseases/chemically induced , Lung Diseases/pathology , Male , Metalloproteases/chemistry , Metalloproteases/toxicity , Mice , Platelet Aggregation/drug effects , Species Specificity , Thrombocytopenia/pathology , Thrombosis/pathology
12.
J Immunol ; 189(7): 3609-17, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22962686

ABSTRACT

There is considerable evidence supporting a role for mold exposure in the pathogenesis and expression of childhood asthma. Aspergillus versicolor and Cladosporium cladosporioides are common molds that have been implicated in asthma. In a model of mold-induced asthma, mice were repeatedly exposed to either A. versicolor or C. cladosporioides spores. The two molds induced distinct phenotypes, and this effect was observed in both BALB/c and C57BL/6 strains. C. cladosporioides induced robust airway hyperresponsiveness (AHR), eosinophilia, and a predominately Th2 response, whereas A. versicolor induced a strong Th17 response and neutrophilic inflammation, but very mild AHR. Neutralization of IL-17A resulted in strong AHR and eosinophilic inflammation following A. versicolor exposure. In Dectin-1-deficient mice, A. versicolor exposure resulted in markedly attenuated IL-17A and robust AHR compared with wild-type mice. In contrast, C. cladosporioides induced AHR and eosinophilic inflammation independent of IL-17A and Dectin-1. A. versicolor, but not C. cladosporioides, spores had increased exposure of ß-glucans on their surface and were able to bind Dectin-1. Thus, the host response to C. cladosporioides was IL-17A- and Dectin-1-independent, whereas Dectin-1- and IL-17A-dependent pathways were protective against the development of asthma after exposure to A. versicolor.


Subject(s)
Anti-Asthmatic Agents/administration & dosage , Aspergillus/immunology , Asthma/immunology , Asthma/pathology , Cladosporium/immunology , Interleukin-17/administration & dosage , Lectins, C-Type/administration & dosage , beta-Glucans/administration & dosage , Animals , Anti-Asthmatic Agents/metabolism , Aspergillus/metabolism , Asthma/prevention & control , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/pathology , Bronchial Hyperreactivity/prevention & control , Cladosporium/metabolism , Eosinophils/immunology , Eosinophils/pathology , Immunophenotyping , Inflammation Mediators/administration & dosage , Lectins, C-Type/deficiency , Lectins, C-Type/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Neutrophils/pathology , Spores, Fungal/immunology , Spores, Fungal/metabolism , Surface Properties , beta-Glucans/metabolism
13.
J Immunol ; 189(1): 312-7, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22649195

ABSTRACT

Glucan particles (GPs) are Saccharomyces cerevisiae cell walls chemically extracted so they are composed primarily of particulate ß-1,3-D-glucans. GPs are recognized by Dectin-1 and are potent complement activators. Mice immunized with Ag-loaded GPs develop robust Ab and CD4(+) T cell responses. In this study, we examined the relative contributions of Dectin-1 and complement to GP phagocytosis and Ag-specific responses to immunization with OVA encapsulated in GPs. The in vitro phagocytosis of GPs by bone marrow-derived dendritic cells was facilitated by heat-labile serum component(s) independently of Dectin-1. This enhanced uptake was not seen with serum from complement component 3 knockout (C3(-/-)) mice and was also inhibited by blocking Abs directed against complement receptor 3. After i.p. injection, percent phagocytosis of GPs by peritoneal macrophages was comparable in wild-type and Dectin-1(-/-) mice and was not inhibited by the soluble ß-glucan antagonist laminarin. In contrast, a much lower percentage of peritoneal macrophages from C3(-/-) mice phagocytosed GPs, and this percentage was further reduced in the presence of laminarin. Subcutaneous immunization of wild-type, Dectin-1(-/-), and C3(-/-) mice with GP-OVA resulted in similar Ag-specific IgG(1) and IgG(2c) type Ab and CD4(+) T cell lymphoproliferative responses. Moreover, while CD4(+) Th1 and Th2 responses measured by ELISPOT assay were similar in the three mouse strains, Th17 responses were reduced in C3(-/-) mice. Thus, although Dectin-1 is necessary for optimal phagocytosis of GPs in the absence of complement, complement dominates when both an intact complement system and Dectin-1 are present. In addition, Th-skewing after GP-based immunization was altered in C3(-/-) mice.


Subject(s)
Complement C3/physiology , Lectins, C-Type/physiology , beta-Glucans/immunology , Animals , Antibodies, Blocking/physiology , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Complement C3/antagonists & inhibitors , Complement C3/deficiency , Dendritic Cells/immunology , Dendritic Cells/metabolism , Lectins, C-Type/administration & dosage , Lectins, C-Type/metabolism , Ligands , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/administration & dosage , Ovalbumin/immunology , Phagocytosis/immunology , beta-Glucans/administration & dosage , beta-Glucans/metabolism
14.
Int Immunopharmacol ; 11(10): 1546-56, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21621644

ABSTRACT

Considerable efforts are currently focused on the biology of DC in view of their possible clinical use as adjuvant for the generation of antigen-specific immunity and lifelong immunologic memory or for the treatment of tumors. We assessed the role of Nattectin a C-type lectin identified in the Thalassophryne nattereri fish venom in DC maturation. Nattectin induced a significant neutrophilic recruitment into peritoneal cavity of mice, followed by macrophages, with lipidic mediators and IL-12 p70 synthesis. Macrophages derived from 7day-Nattectin mice were CD11c+CD11b(low)Ly6(high)F4/80R(high) and express high levels of MHC class II and CD80 molecules. Culture of peritoneal exudates derived macrophages from 7day Nattectin-mice and immature BMDCs with Nattectin markedly increased the surface expression of CD40, CD80, CD86, and MHC class II in a dose-dependent manner, and the production of MMP-2 and MMP-9 distributed in nucleus and cytoplasm of cells, that was associated with strong activity in the culture supernatant. Nattectin treated DCs secreted IL-12 p70 and IL-10. The Nattectin-treated BMDC or macrophage-derived DCs were highly efficient at Ag capture. The specific immune response elicited by Nattectin was characterized by the production of specific antibodies IgG1 and mainly IgG2a with IL-10 and IFN-γ synthesis by splenic cells. These results enable us to address that Nattectin induces the recruitment of Ly6C(high) monocytes into the peritoneum, which exhibit a pro-inflammatory profile, where they differentiate into proliferating F4/80R(high) macrophages. Macrophage-derived DCs mature in the presence of the cytokine milieu generated against Nattectin, exhibiting T cell co-stimulatory molecule expression and induced a Th1 polarized response.


Subject(s)
Batrachoidiformes , Cytokines/metabolism , Dendritic Cells/drug effects , Lectins, C-Type/administration & dosage , Macrophages/drug effects , Animals , Antigen Presentation/drug effects , Antigens, Differentiation/metabolism , Batrachoidiformes/immunology , Cell Transdifferentiation/drug effects , Cells, Cultured , Cellular Microenvironment/drug effects , Cellular Microenvironment/immunology , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Fish Proteins/administration & dosage , Immunity, Cellular/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Th1 Cells/immunology , Th1-Th2 Balance/drug effects
15.
Biochimie ; 93(6): 971-80, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21396978

ABSTRACT

Lectins are glycan-binding receptors that recognize glycan epitopes on foreign pathogens and in the host systems. They can be involved in functions that include innate immunity, development, immune regulation and homeostasis. Several lectins have been purified and characterized from fish species. In this work, using cation-exchange chromatography, a galactose-specific lectin belonging to the family of C-type lectins was isolated from the venom of the Brazilian venomous fish Thalassophryne nattereri. Nattectin is a basic, non-glycosilated, 15 kDa monomeric protein. It exhibits hemagglutination activity that is independent of Ca(2+). We also demonstrated a lectin activity for Nattectin in the innate immune system, especially in neutrophil mobilization in mice, indicating that marine organisms are source of immunomodulator agents.


Subject(s)
Batrachoidiformes , Fish Venoms/metabolism , Immunologic Factors/metabolism , Lectins, C-Type/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Calcium/metabolism , Cell Movement , Conserved Sequence , Fish Venoms/administration & dosage , Fish Venoms/chemistry , Fish Venoms/isolation & purification , Galactose/metabolism , Hemagglutination Tests , Hindlimb/pathology , Humans , Immunity, Innate , Immunologic Factors/administration & dosage , Immunologic Factors/chemistry , Immunologic Factors/isolation & purification , Inflammation/chemically induced , Inflammation/immunology , Lectins, C-Type/administration & dosage , Lectins, C-Type/chemistry , Lectins, C-Type/isolation & purification , Leukocytes/drug effects , Leukocytes/physiology , Matrix Metalloproteinases/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Sequence Analysis, Protein , Structural Homology, Protein
17.
J Control Release ; 151(2): 155-61, 2011 Apr 30.
Article in English | MEDLINE | ID: mdl-21281680

ABSTRACT

Antisense therapy, the first concept of oligonucleotide therapeutics, was proposed more than two decades ago. However, the lack of suitable delivering carriers continues to be a major obstacle to practical therapy. In this study, we present a novel complex consisting of ß-1,3-glucan and antisense oligonucleotide (AS-ODN) as a new candidate of the carriers. We used schizophyllan (SPG) as a ß-1,3-glucan and an AS-ODN sequence to suppress tumor necrosis factor alpha (TNF-α), where the AS-ODN has a (dA)(60) tail to induce complex with SPG. When the complexes were applied to peritoneal macrophages, they were incorporated into the cells via dectin-1 (a ß-1,3-glucan receptor expressed on antigen presenting cells) and suppressed lipopolysaccharide (LPS)-induced TNF-α secretion. In-vivo, AS-ODN/SPG decreased the secretion of TNF-α in serum and drastically reduced the inflammation of LPS-induced hepatitis. This new complex could overcome the long outstanding problem for antisense therapy because of its complexation ability, non-toxicity and high target specificity.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Drug Delivery Systems/methods , Lectins, C-Type/administration & dosage , Oligonucleotides, Antisense/administration & dosage , Tumor Necrosis Factor-alpha/antagonists & inhibitors , beta-Glucans/administration & dosage , Animals , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Hepatitis, Viral, Animal/metabolism , Hepatitis, Viral, Animal/prevention & control , Lectins, C-Type/physiology , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Oligonucleotides, Antisense/metabolism , Protective Agents/administration & dosage , Protective Agents/pharmacology , Tumor Necrosis Factor-alpha/blood , beta-Glucans/chemistry , beta-Glucans/metabolism
18.
Biomaterials ; 32(11): 3094-105, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21262534

ABSTRACT

Dendritic-cell (DC) targeted antigen delivery systems hold promise for enhancing vaccine efficacy and delivery of therapeutics. However, it is not known how the number and density of targeting ligands on such systems may affect DC function and subsequent T cell response. We modified the surface of biodegradable nanoparticles loaded with antigen with different densities of the mAb to the DC lectin DEC-205 receptor and assessed changes in the cytokine response of DCs and T cells. DEC-205 targeted nanoparticles unexpectedly induced a differential cytokine response that depended on the density of ligands on the surface. Strikingly, nanoparticle surface density of DEC-205 mAb increased the amount of anti-inflammatory, IL-10, produced by DCs and T cells. Boosting mice with DEC-205 targeted OVA-nanoparticles after immunization with an antigen in CFA induced a similar pattern of IL-10 response. The correlation between DC production of IL-10 as a function of the density of anti-DEC-205 is shown to be due to cross-linking of the DEC-205 receptor. Cross-linking also increased DC expression of the scavenger receptor CD36, and blockade of CD36 largely abrogated the IL-10 response. Our studies highlight the importance of target ligand density in the design of vaccine delivery systems.


Subject(s)
Antigens, CD/immunology , Dendritic Cells/immunology , Lectins, C-Type/immunology , Nanoparticles/chemistry , Receptors, Cell Surface/immunology , Vaccines/immunology , Animals , Antigens, CD/administration & dosage , Cytokines/metabolism , Flow Cytometry , Interleukin-10/metabolism , Lectins, C-Type/administration & dosage , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Minor Histocompatibility Antigens , Receptors, Cell Surface/administration & dosage , T-Lymphocytes/metabolism , Vaccines/administration & dosage
19.
J Immunol ; 186(2): 891-900, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21160044

ABSTRACT

Although oral dendritic cells (DCs) were shown to induce cell-mediated immunity, the identity and function of the various oral DC subsets involved in this process is unclear. In this study, we examined the mechanisms used by DCs of the buccal mucosa and of the lining mucosa to elicit immunity. After plasmid DNA immunization, buccally immunized mice generated robust local and systemic CD8(+) T cell responses, whereas lower responses were seen by lining immunization. A delayed Ag presentation was monitored in vivo in both groups; yet, a more efficient presentation was mediated by buccal-derived DCs. Restricting transgene expression to CD11c(+) cells resulted in diminished CD8(+) T cell responses in both oral tissues, suggesting that immune induction is mediated mainly by cross-presentation. We then identified, in addition to the previously characterized Langerhans cells (LCs) and interstitial dendritic cells (iDCs), a third DC subset expressing the CD103(+) molecule, which represents an uncharacterized subset of oral iDCs expressing the langerin receptor (Ln(+)iDCs). Using Langerin-DTR mice, we demonstrated that whereas LCs and Ln(+)iDCs were dispensable for T cell induction in lining-immunized mice, LCs were essential for optimal CD8(+) T cell priming in the buccal mucosa. Buccal LCs, however, failed to directly present Ag to CD8(+) T cells, an activity that was mediated by buccal iDCs and Ln(+)iDCs. Taken together, our findings suggest that the mechanisms engaged by oral DCs to prime T cells vary between oral mucosal tissues, thus emphasizing the complexity of the oral immune network. Furthermore, we found a novel regulatory role for buccal LCs in potentiating CD8(+) T cell responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Lymphocyte Activation/immunology , Mouth Mucosa/cytology , Mouth Mucosa/immunology , Animals , Antigen Presentation/genetics , Antigen Presentation/immunology , Antigens, Surface/administration & dosage , Antigens, Surface/biosynthesis , Antigens, Surface/genetics , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/microbiology , Cytotoxicity, Immunologic/genetics , Dendritic Cells/metabolism , Diphtheria Toxin/administration & dosage , Diphtheria Toxin/genetics , Diphtheria Toxin/immunology , Gene Knock-In Techniques , Gingiva/cytology , Gingiva/immunology , Gingiva/microbiology , Humans , Langerhans Cells/cytology , Langerhans Cells/immunology , Langerhans Cells/microbiology , Lectins, C-Type/administration & dosage , Lectins, C-Type/biosynthesis , Lectins, C-Type/genetics , Lymphocyte Activation/genetics , Mannose-Binding Lectins/administration & dosage , Mannose-Binding Lectins/biosynthesis , Mannose-Binding Lectins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Mouth Mucosa/metabolism , Ovalbumin/administration & dosage , Ovalbumin/genetics , Ovalbumin/immunology , Plasmids/administration & dosage , Plasmids/genetics , Plasmids/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology
20.
Anat Rec (Hoboken) ; 294(1): 24-45, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21157914

ABSTRACT

This study was designed to elucidate the potential neuroprotective effects of Reg-2 (regeneration gene protein 2) in a rodent model of spinal cord transection injury at the ninth thoracic level. Reg-2 at 100 and 500 µg, recombinant rat ciliary neurotrophic factor, or vehicle were delivered intrathecally using Alzet miniosmotic pumps. We found that Reg-2 treatment significantly reduced neuronal death in the spinal cord. There was also an attenuation of inflammation at the injury site and an increase in white matter sparing and retained myelination. Retrograde tracing revealed that Reg-2 protected axons of long descending pathways at 6 weeks post-SCI, and the number of FluoroGold-labeled neurons in spinal and supraspinal regions was also significantly increased. Immunofluorescent staining confirmed that the spared white matter contained neurofilament-positive axons. Moreover, behavioral improvements were revealed by Basso Beattie Bresnahan locomotor rating scores and grid-walk analysis. These results suggest that Reg-2 might promote functional recovery by increasing axonal growth, inhibiting neuronal apoptosis, and attenuating spinal cord secondary injury after SCI.


Subject(s)
Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/therapeutic use , Biomarkers, Tumor/administration & dosage , Biomarkers, Tumor/therapeutic use , Lectins, C-Type/administration & dosage , Lectins, C-Type/therapeutic use , Nerve Regeneration/physiology , Neuroprotective Agents/administration & dosage , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/therapy , Animals , Apoptosis Regulatory Proteins/administration & dosage , Apoptosis Regulatory Proteins/therapeutic use , Axons/physiology , Cell Death/physiology , Cervical Vertebrae , Ciliary Neurotrophic Factor/administration & dosage , Disease Models, Animal , Female , Injections, Spinal , Neural Inhibition/physiology , Neuroprotective Agents/therapeutic use , Pancreatitis-Associated Proteins , Random Allocation , Rats , Rats, Sprague-Dawley , Recovery of Function/physiology , Spinal Cord Injuries/physiopathology , Thoracic Vertebrae/injuries
SELECTION OF CITATIONS
SEARCH DETAIL
...