Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
EMBO Mol Med ; 8(4): 422-36, 2016 04 01.
Article in English | MEDLINE | ID: mdl-26976612

ABSTRACT

Influenza virus epidemics potentially cause pneumonia, which is responsible for much of the mortality due to the excessive immune responses. The role of costimulatory OX40-OX40 ligand (OX40L) interactions has been explored in the non-infectious pathology of influenza pneumonia. Here, we describe a critical contribution of OX40L to infectious pathology, with OX40L deficiency, but not OX40 deficiency, resulting in decreased susceptibility to influenza viral infection. Upon infection, bronchiolar progenitors increase in number for repairing the influenza-damaged epithelia. The OX40L expression is induced on the progenitors for the antiviral immunity during the infectious process. However, these defense-like host responses lead to more extensive infection owing to the induced OX40L with α-2,6 sialic acid modification, which augments the interaction with the viral hemagglutinin. In fact, the specific antibody against the sialylated site of OX40L exhibited therapeutic potency in mitigating the OX40L-mediated susceptibility to influenza. Our data illustrate that the influenza-induced expression of OX40L on bronchiolar progenitors has pathogenic value to develop a novel therapeutic approach against influenza.


Subject(s)
Host-Pathogen Interactions , Influenza A virus/physiology , OX40 Ligand/metabolism , Orthomyxoviridae Infections/pathology , Pneumonia, Viral/pathology , Stem Cells/metabolism , Virus Attachment , Animals , Disease Susceptibility , Mice, Inbred C57BL , OX40 Ligand/chemistry , Orthomyxoviridae Infections/virology , Pneumonia, Viral/virology , Protein Processing, Post-Translational , Sialic Acids/analysis , Stem Cells/virology
2.
Plant Cell Rep ; 35(3): 667-79, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26661780

ABSTRACT

KEY MESSAGE: The first Good Manufacturing Practices production of a purification-free rice-based oral cholera vaccine (MucoRice-CTB) from transgenic plants in a closed cultivation system yielded a product meeting regulatory requirements. Despite our knowledge of their advantages, plant-based vaccines remain unavailable for human use in both developing and industrialized countries. A leading, practical obstacle to their widespread use is producing plant-based vaccines that meet governmental regulatory requirements. Here, we report the first production according to current Good Manufacturing Practices of a rice-based vaccine, the cholera vaccine MucoRice-CTB, at an academic institution. To this end, we established specifications and methods for the master seed bank (MSB) of MucoRice-CTB, which was previously generated as a selection-marker-free line, evaluated its propagation, and given that the stored seeds must be renewed periodically. The production of MucoRice-CTB incorporated a closed hydroponic system for cultivating the transgenic plants, to minimize variations in expression and quality during vaccine manufacture. This type of molecular farming factory can be operated year-round, generating three harvests annually, and is cost- and production-effective. Rice was polished to a ratio of 95 % and then powdered to produce the MucoRice-CTB drug substance, and the identity, potency, and safety of the MucoRice-CTB product met pre-established release requirements. The formulation of MucoRice-CTB made by fine-powdering of drug substance and packaged in an aluminum pouch is being evaluated in a physician-initiated phase I study.


Subject(s)
Cholera Vaccines/genetics , Oryza/genetics , Plants, Genetically Modified/genetics , Technology, Pharmaceutical/methods , Administration, Oral , Animals , Blotting, Western , Cholera/immunology , Cholera/microbiology , Cholera/prevention & control , Cholera Toxin/toxicity , Cholera Vaccines/administration & dosage , Cholera Vaccines/immunology , Cost-Benefit Analysis , Diarrhea/chemically induced , Diarrhea/immunology , Diarrhea/prevention & control , Drug Packaging , Drug Stability , Humans , Immunization/methods , Mice , Oryza/growth & development , Plants, Genetically Modified/growth & development , Powders , Reproducibility of Results , Technology, Pharmaceutical/economics , Vibrio cholerae/immunology
3.
PLoS One ; 10(5): e0126849, 2015.
Article in English | MEDLINE | ID: mdl-25978818

ABSTRACT

A lipopolysaccharide from Pantoea agglomerans (LPSpa) has been applied to various fields for human use as a Toll-like receptor 4 ligand and its safety has been confirmed. Here, we showed for the first time the application of LPSpa as an effective mucosal adjuvant for activating vaccine-induced antigen specific immune responses. Mice sublingually immunized with influenza vaccine (HA split vaccine) with LPSpa induced both HA-specific IgG (systemic) and IgA (mucosal) antibody responses, which led to a significant increase in survival rate against lethal influenza virus challenge compared with subcutaneous vaccination. After sublingual administration of ovalbumin with LPSpa, ovalbumin-specific mucosal IgA responses were induced at both mucosal surfaces close to the immunized site and at remote mucosal surfaces. Sublingual administration of LPSpa evoked local antigen-uptake by dendritic cells in cervical lymph nodes. LPSpa induced cytokine production and the maturation and proliferation of innate immune cells via Toll-like receptor 4 in dendritic cells. Collectively, these results suggest that LPSpa can be used as an effective mucosal adjuvant to stimulate and activate local innate immune cells to improve and enhance mucosal vaccine potency against various pathogens.


Subject(s)
Adjuvants, Immunologic/pharmacology , Immunity, Humoral/drug effects , Immunity, Mucosal/drug effects , Influenza Vaccines/immunology , Lipopolysaccharides/pharmacology , Pantoea/immunology , Toll-Like Receptor 4/physiology , Adjuvants, Immunologic/administration & dosage , Administration, Sublingual , Animals , Enzyme-Linked Immunosorbent Assay , Female , Immunity, Humoral/immunology , Immunity, Mucosal/immunology , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/administration & dosage , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
4.
J Vet Med Sci ; 75(3): 395-7, 2013.
Article in English | MEDLINE | ID: mdl-23123943

ABSTRACT

GM1 gangliosidosis is a fatal, progressive neurodegenerative lysosomal storage disease caused by mutations in the ß-galactosidase (GLB1) gene. In feline GM1 gangliosidosis, a pathogenic mutation (c.1448G>C) in the feline GLB1 gene was identified in Siamese cats in the United States and Japan and in Korat cats in Western countries. The present study found the homozygous c.1448G>C mutation in 2 apparent littermate native kittens in Bangladesh that were exhibiting neurological signs. This is the first identification of GM1 gangliosidosis in native domestic cats in Southeast Asia. This pathogenic mutation seems to have been present in the domestic cat population in the Siamese region and may have been transferred to pure breeds such as Siamese and Korat cats originating in this region.


Subject(s)
Amino Acid Substitution , Cat Diseases/enzymology , Gangliosidosis, GM1/veterinary , Gene Expression Regulation, Enzymologic/physiology , beta-Galactosidase/metabolism , Animals , Bangladesh/epidemiology , Cat Diseases/genetics , Cat Diseases/metabolism , Cats , Gangliosidosis, GM1/genetics , Genetic Predisposition to Disease , Mutation , beta-Galactosidase/genetics
5.
Viral Immunol ; 25(5): 433-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22985289

ABSTRACT

Here we examined whether polymeric IgA (pIgA) and monomeric IgA (mIgA) antibodies differ in their ability to neutralize drift viruses within the same subtype. We used an IgA monoclonal antibody (mAb; H1-21) against influenza virus strain A/Hiroshima/52/2005 (A/Hiroshima; H3N2). The mAb was obtained after immunizing mice mucosally with a split-virion (SV) vaccine. The mAb contained both mIgA and pIgA forms. It reacted with the homologous virus and cross-reacted with drift viruses A/New York/55/2004 (H3N2) and A/Wyoming/3/2003 (H3N2) in hemagglutinin-inhibition (HI) and neutralizing Ab assays. The mAb also cross-reacted with A/Panama/2007/99 (H3N2) in an ELISA. We separated the mAb into pIgA and mIgA fractions by gel filtration, and then tested them for neutralizing Ab activity. The neutralizing activity for the A/Hiroshima/52/2005, A/New York/55/2004, and A/Wyoming/3/2003 viruses was lower for the mIgA than the pIgA fraction. However, the neutralizing efficiency for drift variants relative to that for the homotype did not differ between pIgA and mIgA, and pIgA only neutralized variants that could also be neutralized by mIgA. These results suggest that the polymerization of IgA enhances its antiviral immune responses, but does not increase the number of influenza virus strains neutralized by the IgA.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Immunoglobulin A/immunology , Influenza A Virus, H3N2 Subtype/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Cross Reactions/immunology , Female , Influenza Vaccines , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Pertussis Toxin/immunology , Sequence Analysis, RNA
6.
J Feline Med Surg ; 14(12): 900-2, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22772479

ABSTRACT

GM1 gangliosidosis is a fatal, progressive neurodegenerative lysosomal storage disease caused by mutations of the ß-galactosidase (GLB1) gene. In feline GM1 gangliosidosis, a pathogenic mutation (c.1448G>C) of the feline GLB1 gene was identified in Siamese and Korat cats previously diagnosed with the disease in the USA and Italy, respectively. The present study demonstrated the same mutation in a Siamese cat that had been diagnosed with GM1 gangliosidosis in Japan in the 1960s. The mutation was confirmed using DNA extracted from stored paraffin-embedded brain tissue by a direct sequencing method and a polymerase chain reaction-restriction fragment length polymorphism assay. This pathogenic mutation seems to have been distributed around the world.


Subject(s)
Cat Diseases/genetics , Gangliosidosis, GM1/veterinary , beta-Galactosidase/genetics , Animals , Cats , DNA Mutational Analysis/veterinary , Gene Frequency , Mutation
7.
Clin Vaccine Immunol ; 19(7): 979-90, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22552600

ABSTRACT

The antigenicity of seasonal human influenza virus changes continuously; thus, a cross-protective influenza vaccine design needs to be established. Intranasal immunization with an influenza split-virion (SV) vaccine and a mucosal adjuvant induces cross-protection; however, no mucosal adjuvant has been assessed clinically. Formalin-inactivated intact human and avian viruses alone (without adjuvant) induce cross-protection against the highly pathogenic H5N1 avian influenza virus. However, it is unknown whether seasonal human influenza formalin-inactivated whole-virion (WV) vaccine alone induces cross-protection against strains within a subtype or in a different subtype of human influenza virus. Furthermore, there are few reports comparing the cross-protective efficacy of the WV vaccine and SV vaccine-mucosal adjuvant mixtures. Here, we found that the intranasal human influenza WV vaccine alone induced both the innate immune response and acquired immune response, resulting in cross-protection against drift variants within a subtype of human influenza virus. The cross-protective efficacy conferred by the WV vaccine in intranasally immunized mice was almost the same as that conferred by a mixture of SV vaccine and adjuvants. The level of cross-protective efficacy was correlated with the cross-reactive neutralizing antibody titer in the nasal wash and bronchoalveolar fluids. However, neither the SV vaccine with adjuvant nor the WV vaccine induced cross-reactive virus-specific cytotoxic T-lymphocyte activity. These results suggest that the intranasal human WV vaccine injection alone is effective against variants within a virus subtype, mainly through a humoral immune response, and that the cross-protection elicited by the WV vaccine and the SV vaccine plus mucosal adjuvants is similar.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cross Protection , Influenza A virus/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Administration, Intranasal , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Bronchoalveolar Lavage Fluid/immunology , Female , Lung/immunology , Mice , Mice, Inbred BALB C , Nasal Mucosa/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
8.
J Med Virol ; 84(2): 336-44, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22170556

ABSTRACT

The levels and properties of neutralizing antibodies in nasal wash and serum collected from five healthy adults were examined after intranasal administration of an A/Uruguay/716/2007 (H3N2) split vaccine (45 µg hemagglutinin (HA) per dose; five doses, with an interval of 3 weeks between each dose). Prior to the assays, nasal wash samples were concentrated so that the total amount of antibodies was equivalent to about 1/10 of that found in the natural nasal mucus. Vaccination induced virus-specific neutralizing antibody responses, which increased with the number of vaccine doses given. Neutralizing antibodies were produced more efficiently in the nasal passages than in the serum: A ≥4-fold increase in nasal neutralization titres was observed after the second vaccination in four out of five subjects, whereas a rise in serum neutralization titres was observed only after the fifth vaccination. Nasal and serum neutralizing antibodies were mainly found in the polymeric IgA and monomeric IgG fractions, respectively, after gel filtration. Taken together, these results suggest that intranasal administration of an inactivated split vaccine induces high levels of nasal neutralizing antibodies (primarily polymeric IgA) and low levels of serum neutralizing antibodies (primarily monomeric IgG).


Subject(s)
Antibodies, Neutralizing/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Administration, Intranasal , Adult , Aged , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Antibodies, Viral/immunology , Humans , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Influenza, Human/immunology , Male , Middle Aged , Nasal Lavage Fluid/immunology , Vaccination/methods , Young Adult
9.
Vaccine ; 29(9): 1754-61, 2011 Feb 17.
Article in English | MEDLINE | ID: mdl-21219977

ABSTRACT

Effective pneumococcal vaccines are required for preventing secondary bacterial pneumonia, a life-threatening condition, during epidemics of influenza. We examined whether nasal administration of a low dose of pneumococcal surface protein A (PspA) plus polyinosinic-polycytidylic acid (poly(I:C)) could protect against a fatal secondary pneumococcal pneumonia after influenza A virus infection in mice. PspA-specific IgG but not IgA level was higher in the airways and blood of mice nasally administered a low dose of PspA plus poly(I:C) than in mice nasally administered PspA alone or poly(I:C) alone. Binding of PspA-specific IgG increased C3 deposition on the bacterial surface. The survival rate during secondary infection was higher in mice immunized with PspA plus poly(I:C) than in mice immunized with poly(I:C) alone. The significant reduction in bacterial density in the lung and blood was associated with increased survival of immunized mice with secondary pneumonia. Passive transfer of sera from mice immunized with PspA plus poly(I:C) increased the survival of mice infected with secondary pneumonia. Our data suggest that an intranasal PspA vaccine has promising protective effects against secondary pneumonia after influenza and that PspA-specific IgG plays a critical role in this protection.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Bacterial Proteins/administration & dosage , Bacterial Proteins/immunology , Influenza, Human/prevention & control , Pneumococcal Vaccines/administration & dosage , Pneumonia, Pneumococcal/prevention & control , Poly I-C/administration & dosage , Secondary Prevention/methods , Adjuvants, Immunologic/therapeutic use , Administration, Intranasal , Animals , Humans , Immunization, Secondary/methods , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/complications , Influenza, Human/immunology , Mice , Mice, Inbred C57BL , Pneumococcal Infections/etiology , Pneumococcal Infections/immunology , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/immunology , Pneumonia, Pneumococcal/etiology , Pneumonia, Pneumococcal/immunology , Poly I-C/immunology , Poly I-C/therapeutic use
10.
CNS Neurosci Ther ; 17(1): 66-79, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20370806

ABSTRACT

The aim of the present study was to determine whether acetazolamide (AZ) contributes to the inhibition of the fast inactivating transient K(+) current (I(A) ) in adult rat nodose ganglion (NG) neurons. We have previously shown that pretreatment with either AZ or 4-AP attenuated or blocked the CO(2) -induced inhibition of slowly adapting pulmonary stretch receptors in in vivo experiments. The patch-clamp experiments were performed by using the isolated NG neurons. In addition to this, the RT-PCR of mRNA and the expression of voltage-gated K(+) (Kv) 1.4, Kv 4.1, Kv 4.2, and Kv 4.3 channel proteins from nodose ganglia were examined. We used NG neurons sensitive to the 1 mM AZ application. The application of 1 mM AZ inhibited the I(A) by approximately 27% and the additional application of 4-AP (1 mM) further inhibited I(A) by 48%. The application of 0.1 µM α-dendrotoxin (α-DTX), a slow inactivating transient K(+) current (I(D) ) blocker, inhibited the baseline I(A) by approximately 27%, and the additional application of 1 mM AZ further decreased the I(A) by 51%. In current clamp experiments, AZ application (1 mM) increased the number of action potentials due to the decreased duration of the depolarizing phase of action potentials and/or due to a reduction in the resting membrane potential. Four voltage-gated K(+) channel proteins were present, and most (80-90%) of the four Kv channels immunoreactive neurons showed the co-expression of carbonic anhydrase-II (CA-II) immunoreactivity. These results indicate that the application of AZ causes the reduction in I(A) via the inhibition of four voltage-gated K(+) channel (Kv) proteins without affecting I(D).


Subject(s)
Acetazolamide/pharmacology , Action Potentials/drug effects , Neurons/drug effects , Nodose Ganglion/drug effects , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/drug effects , 4-Aminopyridine/pharmacology , Animals , Carbonic Anhydrase Inhibitors/pharmacology , Cells, Cultured , Drug Interactions , Male , Neurons/classification , Nodose Ganglion/cytology , Patch-Clamp Techniques , Potassium Channels, Voltage-Gated/physiology , Protein Subunits , Rats , Rats, Wistar
11.
Sci Transl Med ; 2(25): 25ra24, 2010 Mar 31.
Article in English | MEDLINE | ID: mdl-20424013

ABSTRACT

A variety of different vaccine types are available for H1N1 influenza A virus infections; however, their immunological mechanisms of action remain unclear. Here, we show that plasmacytoid dendritic cells (pDCs) and type I interferon (IFN)-mediated signaling delineate the immunogenicity of live attenuated virus, inactivated whole-virus (WV), and split-virus vaccines. Although Toll-like receptor 7 acted as the adjuvant receptor for the immunogenicity of both live virus and WV vaccines, the requirement for type I IFN production by pDCs for the immunogenicity of the vaccines was restricted to WV. A split vaccine commonly used in humans failed to immunize naïve mice, but a pDC-activating adjuvant could restore immunogenicity. In blood from human adults, however, split vaccine alone could recall memory T cell responses, underscoring the importance of this adjuvant pathway for primary, but not secondary, vaccination.


Subject(s)
Dendritic Cells/immunology , Influenza Vaccines/immunology , Adult , Animals , Female , Flow Cytometry , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/classification , Male , Mice , Mice, Mutant Strains , Middle Aged , Toll-Like Receptor 7/immunology , Vaccines, Attenuated/immunology
12.
Vaccine ; 27(42): 5896-905, 2009 Sep 25.
Article in English | MEDLINE | ID: mdl-19647814

ABSTRACT

Adding poly(gamma-glutamic acid) nano-particles (gamma-PGA-NPs), a safe, natural material, to subcutaneous immunization with influenza virus hemagglutinin (HA) vaccine increases the protective immune responses against influenza virus in mice. Here, we examined whether intranasal administration of the HA vaccine with gamma-PGA-NPs would induce protection from influenza virus challenge in mice. Intranasal immunization with the mixture of gamma-PGA-NPs and HA vaccine from an influenza virus strain A/PR/8/34 (H1N1) or A/New Caledonia/20/99 (H1N1) enhanced protection of mice from A/PR/8/34 infection. Intranasal immunization with A/New Caledonia/20/99 HA vaccine and gamma-PGA-NPs induced cell-mediated immune responses and neutralizing antibody production for both A/New Caledonia/20/99 and A/PR/8/34. These data suggest that gamma-PGA-NPs may have potential for clinical applications as a mucosal adjuvant.


Subject(s)
Adjuvants, Immunologic/pharmacology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/immunology , Nanoparticles , Orthomyxoviridae Infections/prevention & control , Polyglutamic Acid/analogs & derivatives , Administration, Intranasal , Animals , Antibodies, Viral/immunology , Cell Proliferation , Cytokines/immunology , Female , Immunity, Cellular , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Mice , Mice, Inbred BALB C , Neutralization Tests , Polyglutamic Acid/immunology , Spleen/cytology , Spleen/immunology
13.
J Infect Dis ; 198(9): 1327-33, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18774884

ABSTRACT

Cell-mediated immunity (CMI) is critical for the prevention and control of varicella-zoster virus (VZV)-related disease. To assess CMI to VZV, a varicella skin test and interferon-gamma enzyme-linked immunospot (ELISPOT) assay were both performed in healthy volunteers, and the results were compared. A total of 151 subjects were examined: 16 aged 20-29 years, 26 aged 30-39 years, 18 aged 40-49 years, 73 aged 50-59 years, and 18 aged 60-69 years. All were seropositive by a glycoprotein antigen-based enzyme-linked immunosorbent assay (gpELISA). Skin test reactivity was significantly correlated with the ELISPOT count, and both decreased with increasing age, indicating an age-dependent decline in CMI to VZV. In contrast, the antibody titer obtained by the gpELISA did not correlate with skin test reactivity. The results suggest that the skin test and ELISPOT assay are both reliable for assessing CMI to VZV and can easily be applied to screen individuals susceptible to the development of herpes zoster.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Herpesvirus 3, Human/immunology , Immunity, Cellular/physiology , Interferon-gamma/immunology , Adult , Aged , Antibodies, Viral/blood , Antibodies, Viral/immunology , Female , Humans , Male , Middle Aged , Skin Tests
14.
Eur J Pain ; 12(2): 189-95, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17584507

ABSTRACT

Voltage-gated K+ (Kv) channels are one of the important physiological regulators of the membrane potentials in excitable cells, including sensory ganglion neurons. The aim of the present study was to investigate whether temporomandibular joint (TMJ) inflammation alters expression of Kv channel subtype 1.4 (Kv1.4) of trigeminal ganglion (TRG) neurons innervating TMJ relating allodynia (pain caused by normally innoxious stimulation), by using both behavioral and immunohistochemical techniques. TMJ inflammation was induced by injection of Complete Freund's Adjuvant (CFA) into the rat TMJ. The threshold for escape from mechanical stimulation applied to the orofacial area in TMJ inflamed rats was significantly lower than that in naïve rats. TMJ afferents were identified by fluorogold (FG) labeling. The mean numbers of Kv1.4-/neurofilament (NF) 200(myelinated fiber marker) positive- and negative-immunoreactivities FG-labeled small-/medium-diameter TRG neurons in inflamed rats were significantly decreased when compared with those in the naïve rats. These findings suggest that TMJ inflammation reduces the expression of Kv1.4 subunits in the small-/medium sized (Adelta-/C-) TRG neurons and this may contribute to trigeminal inflammatory allodynia in TMJ disorder. These results lead us to suggest that Kv channel openers may be a potential therapeutic agents for prevention of mechanical allodynia.


Subject(s)
Arthritis/metabolism , Kv1.4 Potassium Channel/metabolism , Neurons/metabolism , Temporomandibular Joint , Trigeminal Ganglion/metabolism , Animals , Arthritis/chemically induced , Arthritis/pathology , Cell Size , Freund's Adjuvant , Hyperalgesia/chemically induced , Immunohistochemistry , Male , Rats , Rats, Wistar , Temporomandibular Joint/innervation , Trigeminal Ganglion/pathology
15.
J Immunol ; 179(7): 4711-20, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17878370

ABSTRACT

The innate immune system recognizes influenza A virus via TLR 7 or retinoic acid-inducible gene I in a cell-type specific manner in vitro, however, physiological function(s) of the MyD88- or interferon-beta promoter stimulator 1 (IPS-1)-dependent signaling pathways in antiviral responses in vivo remain unclear. In this study, we show that although either MyD88- or IPS-1-signaling pathway was sufficient to control initial antiviral responses to intranasal influenza A virus infection, mice lacking both pathways failed to show antiviral responses, resulting in increased viral load in the lung. By contrast, induction of B cells or CD4 T cells specific to the dominant hemagglutinin or nuclear protein Ags respectively, was strictly dependent on MyD88 signaling, but not IPS-1 signaling, whereas induction of nuclear protein Ag-specific CD8 T cells was not impaired in the absence of either MyD88 or IPS-1. Moreover, vaccination of TLR7- and MyD88-deficient mice with inactivated virus failed to confer protection against a lethal live virus challenge. These results strongly suggest that either the MyD88 or IPS-1 signaling pathway is sufficient for initial antiviral responses, whereas the protective adaptive immune responses to influenza A virus are governed by the TLR7-MyD88 pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Influenza A virus/immunology , Orthomyxoviridae Infections/immunology , Signal Transduction/immunology , Toll-Like Receptors/immunology , Vaccination , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigens/immunology , Cells, Cultured , Immunity, Innate/immunology , Interferon Type I/biosynthesis , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Orthomyxoviridae Infections/metabolism , Receptors, Cell Surface , Toll-Like Receptors/deficiency , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
16.
Neuropharmacology ; 52(3): 904-24, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17140607

ABSTRACT

We examined whether 8-bromo-cAMP (8-Br-cAMP)-induced modification of tetrodotoxin-resistant (TTX-R) sodium current in neonatal rat nodose ganglion neurons is mediated by the activation of protein kinase A (PKA) and/or protein kinase C (PKC). In 8-Br-cAMP applications ranging from 0.001 to 1.0mM, 8-Br-cAMP at 0.1mM showed a maximal increase in the peak TTX-R Na(+) (Nav1.8) current and produced a hyperpolarizing shift in the conductance-voltage (G-V) curve. The PKC inhibitor bisindolylmaleimide Ro-31-8425 (Ro-31-8425, 0.5microM) decreased the peak Nav 1.8 current. The Ro-31-8425-induced modulation of the G(V)(1/2) baseline (a percent change in G at baseline V1/2) was not affected by additional 8-Br-cAMP application (0.1mM). The maximal increase in Nav 1.8 currents was seen at 0.1microM after the application of a PKC activator, phorbol 12-myristate 13-acetate (PMA) and forskolin. The PMA-induced increase in Nav 1.8 currents was not significantly affected by additional 0.1mM 8-Br-cAMP application. Intracellular application of a PKA inhibitor, protein kinase inhibitor (PKI, 0.01mM), inhibited the baseline Nav 1.8 current, significantly attenuated the 8-Br-cAMP-and PMA-induced increase in the peak Nav 1.8 current, and caused a significant increase in the slope factor of the inactivation curve. The PKI application at a higher concentration (0.5mM) greatly inhibited the PMA (0.1microM)-induced increase in the peak Nav 1.8 current amplitude and further enhanced the Ro-31-8425-induced decrease in the current. These results suggest that the 8-Br-cAMP-induced increase in Nav 1.8 currents may be mediated by activation of both PKA and PKC.


Subject(s)
8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Nerve Tissue Proteins/physiology , Neurons/drug effects , Nodose Ganglion/cytology , Sodium Channel Blockers/pharmacology , Sodium Channels/physiology , Tetrodotoxin/pharmacology , Animals , Animals, Newborn , Cells, Cultured , Colforsin/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Gene Expression/physiology , Membrane Potentials/drug effects , NAV1.8 Voltage-Gated Sodium Channel , Neural Inhibition/drug effects , Neural Inhibition/radiation effects , Neurons/cytology , Patch-Clamp Techniques , RNA, Messenger/biosynthesis , Rats , Reverse Transcriptase Polymerase Chain Reaction/methods
17.
Pain ; 129(1-2): 155-66, 2007 May.
Article in English | MEDLINE | ID: mdl-17127002

ABSTRACT

Peripheral nerve injury activates satellite cells to produce interleukin 1beta (IL-1beta) which mediates inflammation and hyperalgesia. This study investigated the hypothesis that activation of satellite glial cells modulates the excitability of trigeminal ganglion (TRG) neurons via IL-1beta following inflammation. Inflammation was induced by injection of complete Freund's adjuvant (CFA) into the whisker pad area. The threshold for escape from mechanical stimulation applied to the whisker pad in inflamed rats was significantly lower than that in control. Two days post-CFA injection, the mean percentage of TRG neurons encircled by glial fibrillary acidic protein (GFAP)-/IL-1beta-immunoreactive cells was significantly increased compared to controls. GFAP and IL-1beta immunoreactivities were coexpressed in the same cells. Fluorogold (FG) labeling identified the site of inflammation. The number of FG-labeled IL-receptor type I (IL-1RI) TRG neurons in inflamed rats was significantly greater than in controls. In FG-labeled small TRG neurons, the size of IL-1beta (1 nM) induced-depolarization in inflamed rats was larger than in controls. IL-1beta application significantly increased firing rates evoked by depolarizing pulses in the neurons of inflamed rats, compared to controls. The response to IL-1beta was abolished by treatment with the IL-1RI antagonist. These results suggest that activation of satellite glial cells modulates the excitability of small-diameter TRG neurons via IL-1beta following inflammation, and that the upregulation of IL-1RI in the soma may contribute to the mechanism underlying inflammatory hyperalgesia. Therefore IL-1beta blockers are potential therapeutic agents for prevention of trigeminal hyperalgesia.


Subject(s)
Cytokines/pharmacology , Neuroglia/chemistry , Neurons/physiology , Peripheral Nervous System Diseases/pathology , Peripheral Nervous System Diseases/physiopathology , Trigeminal Ganglion/pathology , Animals , Escape Reaction/drug effects , Escape Reaction/physiology , Freund's Adjuvant , Functional Laterality , Glial Fibrillary Acidic Protein/metabolism , Inflammation/chemically induced , Inflammation/complications , Male , Membrane Potentials/drug effects , Neuroglia/metabolism , Neurons/drug effects , Patch-Clamp Techniques/methods , Peripheral Nervous System Diseases/etiology , Rats , Rats, Wistar , Sensory Thresholds/drug effects , Sensory Thresholds/physiology , Skin/innervation , Stilbamidines/metabolism
18.
Odontology ; 94(1): 22-8, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16998614

ABSTRACT

The aim of the present study was to determine whether there is a convergence of inputs from tooth pulp (TP) and the superior sagittal sinus (SSS) on rat C1 spinal neurons, and to examine the effects of iontophoretically applied N-methyl-D: -aspartate (NMDA) and non-NMDA receptor antagonists on the SSS-evoked activity of C1 neurons. Extracellular single unit-recordings were made from 20 C1 units responding to TP electrical stimulation with a constant temporal relationship to a digastric electromyogram signal, using a multibarrel electrode in pentobarbital-anesthetized rats. Ninety percent of C1 neurons (18/20) responding to TP stimulation also responded to the SSS stimulation. These neurons were considered to be SSS-afferent inputs from Adelta-fibers (5.8 +/- 0.6 m/s; n = 18), based on the calculation of nerve conduction velocity. After the iontophoretic application (30, 50, and 70 nA) of an NMDA receptor blocker (5R-10S)-(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d] cycloheptene-5,10-imine hydrogen maleate (MK801) or a non-NMDA receptor blocker (6-cyano-7-nitroquinoxaline-2,3-dione) (CNQX), the mean number of spikes responding to the SSS stimulation significantly decreased (30, 50, and 70 nA; P < 0.05). These results suggest that there is a convergence of inputs from SSS and TP afferents on C1 neurons; it is possible that both NMDA and non-NMDA receptors located on C1 neurons may be targets for the treatment of the trigeminal referred pain associated with migraine.


Subject(s)
Dental Pulp/innervation , Dura Mater/physiology , Excitatory Amino Acid Antagonists/pharmacology , Pain, Referred/physiopathology , Posterior Horn Cells/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Toothache/physiopathology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Dizocilpine Maleate/pharmacology , Electric Stimulation , Electromyography , Iontophoresis , Male , Neck Muscles/innervation , Nociceptors/drug effects , Nociceptors/physiology , Rats , Rats, Wistar , Trigeminal Neuralgia/physiopathology
19.
Brain Res ; 1109(1): 70-3, 2006 Sep 13.
Article in English | MEDLINE | ID: mdl-16836984

ABSTRACT

The effect of iontophoretic application of the 5-HT3 receptor agonist, phenylbiguanide (PBG), on the excitation of the trigeminal spinal nucleus oralis (TSNO) neurons to tooth-pulp (TP) stimulation was examined. The PBG application inhibited the TP-evoked TSNO neuronal excitation, and this inhibition was completely blocked by co-application of a GABAA receptor antagonist, bicuculline. The results suggest that the activation of 5-HT3 receptors elicits GABA release in the TSNO.


Subject(s)
Dental Pulp/innervation , Interneurons/physiology , Receptors, Serotonin, 5-HT3/physiology , Trigeminal Nucleus, Spinal/cytology , gamma-Aminobutyric Acid/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Bicuculline/pharmacology , Biguanides/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , GABA Antagonists/pharmacology , Indoles/pharmacology , Interneurons/drug effects , Iontophoresis/methods , Male , Rats , Rats, Wistar , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Tropisetron
20.
Life Sci ; 78(1): 112-20, 2005 Nov 19.
Article in English | MEDLINE | ID: mdl-16115651

ABSTRACT

The combined effects of ouabain (Na(+)-K(+) ATPase inhibitor) and hyperinflation (inflation volume=three tidal volumes) on slowly adapting pulmonary stretch receptors (SARs) were studied before and after administration of nifedipine (an L-type Ca(2+) channel blocker) and KB-R7943 (a reverse-mode Na(+)-Ca(2+) exchanger blocker) in anesthetized, artificially ventilated rabbits after bilateral vagotomy. Before ouabain administration, hyperinflation stimulated SAR activity. After 20 min of ouabain administration (30 microg/kg) the SARs increased discharge rates in normal inflation. Under these conditions, hyperinflation initially stimulated SAR activity but subsequently inhibited the activity at peak inflation. Additional administration of 60 microg/kg ouabain (total dose=90 microg/kg) caused a further stimulation of SAR activity, but 20 min later both normal inflation and hyperinflation resulted in a greater inhibition of the receptor activity. The hyperinflation-induced SAR inhibition in the presence of ouabain (30 microg/kg) was not significantly altered by administration of either nifedipine (2 and 4 mg/kg) or KB-R7943 (1 and 3 mg/kg). In another series of experiments, we further examined the combined effects of ouabain and hyperinflation in veratridine (a Na(+) channel opener, 40 microg/kg)-treated animals. After recovery from the veratridine effect on SAR activity, which vigorously stimulated the receptor activity, ouabain treatment (30 microg/kg) that silenced the receptor activity at peak inflation greatly inhibited hyperinflation-induced SAR stimulation. These results suggest that hyperinflation-induced SAR inhibition in the presence of ouabain may be related to a Na(+) overload, but not to a Ca(2+) influx via activation of L-type Ca(2+) channels, in the SAR endings.


Subject(s)
Enzyme Inhibitors/pharmacology , Lung/physiology , Ouabain/pharmacology , Pulmonary Stretch Receptors/drug effects , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Lung/drug effects , Nerve Endings/drug effects , Nifedipine/pharmacology , Rabbits , Respiration, Artificial , Sodium/physiology , Thiourea/analogs & derivatives , Thiourea/pharmacology , Vagotomy , Veratridine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...