Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Mol Pharm ; 19(6): 1814-1824, 2022 06 06.
Article in English | MEDLINE | ID: mdl-35302764

ABSTRACT

Continuous outbreaks of pertussis around the world suggest inadequate immune protection in infants and weakened immune responses induced over time by the acellular pertussis vaccine. Vaccine adjuvants provide a means to improve vaccine immunogenicity and support long-term adaptive immunity against pertussis. An acellular pertussis vaccine was prepared with pertactin, pertussis toxin, and fimbriae 2/3 antigens combined with a triple-adjuvant system consisting of innate defense regulator peptide IDR 1002, a Toll-like receptor-3 agonist poly(I:C), and a polyphosphazene in a fixed combination. The vaccine was delivered intranasally in a cationic lipid nanoparticle formulation fabricated by simple admixture and two schema for addition of antigens (LT-A, antigens associated outside of L-TriAdj, and LAT, antigens associated inside of L-TriAdj) to optimize particle size and cationic surface charge. In the former, antigens were associated with the lipidic formulation of the triple adjuvant by electrostatic attraction. In the latter, the antigens resided in the interior of the lipid nanoparticle. Two dose levels of antigens were used with adjuvant comprised of the triple adjuvant with or without the lipid nanoparticle carrier. Formulation of vaccines with the triple adjuvant stimulated systemic and mucosal immune responses. The lipid nanoparticle vaccines favored a Th1 type of response with higher IgG2a and IgA serum antibody titers particularly for pertussis toxin and pertactin formulated at the 5 µg dose level in the admixed formulation. Additionally, the lipid nanoparticle vaccines resulted in high nasal SIgA antibodies and an early (4 weeks post vaccination) response after a single vaccination dose. The LT-A nanoparticles trended toward higher titers of serum antibodies compared to LAT. The cationic lipid-based vaccine nanoparticles formulated with a triple adjuvant showed encouraging results as a potential formulation for intranasally administered pertussis vaccines.


Subject(s)
Adjuvants, Immunologic , Liposomes , Nanoparticles , Pertussis Vaccine , Whooping Cough , Animals , Antibodies, Bacterial , Bordetella pertussis , Cations , Humans , Liposomes/administration & dosage , Mice , Mice, Inbred BALB C , Nanoparticles/administration & dosage , Pertussis Toxin/administration & dosage , Pertussis Toxin/immunology , Pertussis Vaccine/administration & dosage , Pertussis Vaccine/chemistry , Pertussis Vaccine/immunology , Vaccination , Whooping Cough/prevention & control
2.
Immunology ; 164(3): 602-616, 2021 11.
Article in English | MEDLINE | ID: mdl-34310708

ABSTRACT

Asymmetric dimethylarginine (ADMA) is an endogenous nitric oxide synthase (NOS) inhibitor/uncoupler inducing vascular pathology. Vascular pathology is an important factor for the development and progression of CNS pathology of MS, yet the role of ADMA in MS remains elusive. Patients with multiple sclerosis (MS) are reported to have elevated blood levels of ADMA, and mice with experimental autoimmune encephalomyelitis (EAE, an animal model of MS) generated by auto-immunization of myelin oligodendrocyte glycoprotein (MOG) and blood-brain barrier (BBB) disruption by pertussis toxin also had increased blood ADMA levels in parallel with induction of clinical disease. To explore the role of ADMA in EAE pathogenesis, EAE mice were treated with a daily dose of ADMA. It is of special interest that ADMA treatment enhanced the BBB disruption in EAE mice and exacerbated the clinical and CNS disease of EAE. ADMA treatment also induced the BBB disruption and EAE disease in MOG-immunized mice even without pertussis toxin treatment, suggesting the role of ADMA in BBB dysfunction in EAE. T-cell polarization studies also documented that ADMA treatment promotes TH 1- and TH 17-mediated immune responses but without affecting Treg-mediated immune response in EAE mice as well as in in vitro T-cell culture. Taken together, these data, for the first time, document the vascular and immunopathogenic roles of ADMA in EAE, thus pointing to the potential of ADMA-mediated mechanism as a new target of potential therapy for MS.


Subject(s)
Arginine/analogs & derivatives , Blood-Brain Barrier/pathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Animals , Arginine/metabolism , Blood-Brain Barrier/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Mice , Multiple Sclerosis/pathology , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Myelin-Oligodendrocyte Glycoprotein/immunology , Pertussis Toxin/administration & dosage , Pertussis Toxin/immunology
3.
Int J Mol Sci ; 22(6)2021 Mar 13.
Article in English | MEDLINE | ID: mdl-33805762

ABSTRACT

Pertussis toxin (PTX) is a required co-adjuvant for experimental autoimmune encephalomyelitis (EAE) induced by immunization with myelin antigen. However, PTX's effects on EAE induced by the transfer of myelin-specific T helper cells is not known. Therefore, we investigated how PTX affects the Th17 transfer EAE model (Th17-EAE). We found that PTX significantly reduced Th17-EAE by inhibiting chemokine-receptor-dependent trafficking of Th17 cells. Strikingly, PTX also promoted the accumulation of B cells in the CNS, suggesting that PTX alters the disease toward a B-cell-dependent pathology. To determine the role of B cells, we compared the effects of PTX on Th17-EAE in wild-type (WT) and B-cell-deficient (µMT) mice. Without PTX treatment, disease severity was equivalent between WT and µMT mice. In contrast, with PTX treatment, the µMT mice had significantly less disease and a reduction in pathogenic Th17 cells in the CNS compared to the WT mice. In conclusion, this study shows that PTX inhibits the migration of pathogenic Th17 cells, while promoting the accumulation of pathogenic B cells in the CNS during Th17-EAE. These data provide useful methodological information for adoptive-transfer Th17-EAE and, furthermore, describe another important experimental system to study the pathogenic mechanisms of B cells in multiple sclerosis.


Subject(s)
B-Lymphocytes/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Pertussis Toxin/administration & dosage , Th17 Cells/pathology , Adoptive Transfer/methods , Animals , B-Lymphocytes/immunology , Cell Movement/immunology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/mortality , Female , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Severity of Illness Index , Th17 Cells/immunology , Th17 Cells/transplantation
4.
Clin Microbiol Infect ; 27(6): 878-885, 2021 Jun.
Article in English | MEDLINE | ID: mdl-32896653

ABSTRACT

OBJECTIVES: Protection induced by acellular vaccines can be short, requiring novel immunization strategies. Objectives of this study were to evaluate safety and capacity of a recombinant pertussis toxin (PTgen) -coated Viaskin® epicutaneous patch to recall memory responses in healthy adults. METHODS: This double-blind, placebo-controlled randomized trial (Phase I) assessed the safety and immunogenicity of PTgen administered on days 0 and 14 to healthy adults using Viaskin® patches applied directly or after epidermal laser-based skin preparation. Patch administration was followed by Boostrix®dTpa on day 42. Antibodies were assessed at days 0, 14, 28, 42 and 70. RESULTS: Among 102 volunteers enrolled, 80 received Viaskin-PT (Viaskin-PT 25 µg (n = 25), Viaskin-PT 50 µg (n = 25), laser + Viaskin-PT 25 µg (n = 5), laser + Viaskin-PT 50 µg (n = 25)), Viaskin-placebo (n = 10) or laser + Viaskin-placebo (n = 2). Incidence of adverse events was similar across groups (any local event: 21/25 (84.0%), 24/25 (96.0%), 4/5 (80.0%), 24/25 (96.0%), 8/10 (80.0%), 10/12 (83.0%), respectively). Direct application induced no detectable response. On day 42, PT-IgG geometric mean concentrations were significantly higher following laser + Viaskin-PT 25 µg and 50 µg (139.87 (95% CI 87.30-224.10) and 121.76 (95% CI 95.04-156.00), respectively), than laser + Viaskin-placebo (59.49, 95% CI 39.37-89.90). Seroresponse rates were higher following laser + Viaskin-PT 25 µg (4/5 (80.0%), 95% CI 28.4-99.5) and 50 µg (22/25 (88.0%), 95% CI 68.8-97.5) than laser + Viaskin-placebo (0/12 (0.0%), 95% CI 0.0-26.5). CONCLUSIONS: Viaskin-PT applied after laser-based epidermal skin preparation showed encouraging safety and immunogenicity results: anti-PT booster responses were not inferior to those elicited by Boostrix®dTpa. This study is registered at ClinicalTrials.gov (NCT03035370) and was funded by DBV Technologies.


Subject(s)
Pertussis Toxin/immunology , Administration, Cutaneous , Adolescent , Adult , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Male , Pertussis Toxin/administration & dosage , Young Adult
5.
Int Immunopharmacol ; 80: 106138, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32007705

ABSTRACT

It has been shown that the blockade of chemokine receptor type 5 can dampen inflammatory reaction within the central nervous system (CNS). In the present study, we utilized maraviroc, a potent antagonist o CCR5, to examine whether this drug can mitigate neuroinflammation in the spinal cord of mice induced by experimental autoimmune encephalitis (EAE), considered a murine model of multiple sclerosis (MS). For this aim, mice were immunized with myelin oligodendrocyte glycoprotein 35-55 (MOG35-55), followed by pertussis toxin to induce paralysis in EAE mice. The animals intraperitoneally received various doses of maraviroc (5, 25, and 50 mg/kg body weight) when the early clinical signs of EAE appeared. The results demonstrated that the administration of maraviroc led to a marked decrease in the clinical score and improvement in behavioral motor functions. Moreover, our finding indicated that the administration of maraviroc significantly attenuates the infiltration of inflammatory cells to the spinal cord, microgliosis, astrogliosis, pro-inflammatory cytokines, and cell death in EAE mice. The flow cytometry data indicated that a decreased number of CD4+ and CD8+ T cells in the peripheral blood of mice with EAE without affecting the number of T regulatory cells (CD4 + CD25+ forkhead box protein 3+). Finally, it seems that maraviroc is well-tolerated, and targeting CCR5 could open up a new horizon in the treatment of MS.


Subject(s)
CCR5 Receptor Antagonists/pharmacology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Maraviroc/pharmacology , Multiple Sclerosis/drug therapy , Signal Transduction/drug effects , Animals , CCR5 Receptor Antagonists/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Caspase 3/metabolism , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Lymphocyte Count , Maraviroc/therapeutic use , Mice , Multiple Sclerosis/blood , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Myelin-Oligodendrocyte Glycoprotein/immunology , Pertussis Toxin/administration & dosage , Pertussis Toxin/immunology , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, CCR5/metabolism , Signal Transduction/immunology , Spinal Cord/immunology , Spinal Cord/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , bcl-2-Associated X Protein/metabolism
6.
J Mol Med (Berl) ; 98(1): 97-110, 2020 01.
Article in English | MEDLINE | ID: mdl-31811326

ABSTRACT

Pertussis toxin (PTX) is a potent virulence factor in patients suffering from whooping cough, but in its detoxified version, it is applied for vaccination. It is thought to contribute to the pathology of the disease including various CNS malfunctions. Based on its enzymatic activity, PTX disrupts GPCR-dependent signaling by modifying the α-subunit of heterotrimeric Gi/o-proteins. It is also extensively used as a research tool to study neuronal functions in vivo and in vitro. However, data demonstrating the penetration of PTX from the blood into the brain are missing. Here, we examined the Gαi/o-modifying activity of PTX in murine brains after its parenteral application. Ex vivo biodistribution analysis of [124I]-PTX displayed poor distribution to the brain while relatively high concentrations were visible in the pancreas. PTX affected CNS and endocrine functions of the pancreas as shown by open-field and glucose tolerance tests, respectively. However, while pancreatic islet Gαi/o-proteins were modified, their neuronal counterparts in brain tissue were resistant towards PTX as indicated by different autoradiographic and immunoblot SDS-PAGE analyses. In contrast, PTX easily modified brain Gαi/o-proteins ex vivo. An attempt to increase BBB permeability by application of hypertonic mannitol did not show PTX activity on neuronal G proteins. Consistent with these findings, in vivo MRI analysis did not point to an increased blood-brain barrier (BBB) permeability following PTX treatment. Our data demonstrate that the CNS is protected from PTX. Thus, we hypothesize that the BBB hinders PTX to penetrate into the CNS and to deliver its enzymatic activity to brain Gαi/o-proteins. KEY MESSAGES: i.p. applied PTX is poorly retained in the brain while reaches high concentration in the pancreas. Pancreatic islet Gαi/o- but not cerebral Gαi/o-proteins are modified by i.p. administered PTX. Gαi/o-proteins from isolated cerebral cell membranes were easily modified by PTX ex vivo. CNS is protected from i.p. administered PTX. PTX does not permeabilize the BBB.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Injections/methods , Neuroprotection , Pertussis Toxin/administration & dosage , Pertussis Toxin/metabolism , Signal Transduction/drug effects , Animals , Blood-Brain Barrier/diagnostic imaging , Blood-Brain Barrier/metabolism , Capillary Permeability/drug effects , Cell Membrane/metabolism , Female , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , Iodine Radioisotopes , Islets of Langerhans/diagnostic imaging , Islets of Langerhans/metabolism , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Tissue Distribution
7.
Mol Pharmacol ; 97(1): 2-8, 2020 01.
Article in English | MEDLINE | ID: mdl-31704717

ABSTRACT

The thyrotropin (TSH) receptor (TSHR) signals via G proteins of all four classes and ß-arrestin 1. Stimulation of TSHR leads to increasing cAMP production that has been reported as a monotonic dose-response curve that plateaus at high TSH doses. In HEK 293 cells overexpressing TSHRs (HEK-TSHR cells), we found that TSHR activation exhibits an "inverted U-shaped dose-response curve" with increasing cAMP production at low doses of TSH and decreased cAMP production at high doses (>1 mU/ml). Since protein kinase A inhibition by H-89 and knockdown of ß-arrestin 1 or ß-arrestin 2 did not affect the decreased cAMP production at high TSH doses, we studied the roles of TSHR downregulation and of Gi/Go proteins. A high TSH dose (100 mU/ml) caused a 33% decrease in cell-surface TSHR. However, because inhibiting TSHR downregulation with combined expression of a dominant negative dynamin 1 and ß-arrestin 2 knockdown had no effect, we concluded that downregulation is not involved in the biphasic cAMP response. Pertussis toxin, which inhibits activation of Gi/Go, abolished the biphasic response with no statistically significant difference in cAMP levels at 1 and 100 mU/ml TSH. Concordantly, co-knockdown of Gi/Go proteins increased cAMP levels stimulated by 100 mU/ml TSH from 55% to 73% of the peak level. These data show that biphasic regulation of cAMP production is mediated by Gs and Gi/Go at low and high TSH doses, respectively, which may represent a mechanism to prevent overstimulation in TSHR-expressing cells. SIGNIFICANCE STATEMENT: We demonstrate biphasic regulation of TSH-mediated cAMP production involving coupling of the TSH receptor (TSHR) to Gs at low TSH doses and to Gi/o at high TSH doses. We suggest that this biphasic cAMP response allows the TSHR to mediate responses at lower levels of TSH and that decreased cAMP production at high doses may represent a mechanism to prevent overstimulation of TSHR-expressing cells. This mechanism could prevent chronic stimulation of thyroid gland function.


Subject(s)
Cyclic AMP/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Receptors, Thyrotropin/metabolism , Signal Transduction/drug effects , Thyrotropin/administration & dosage , Dose-Response Relationship, Drug , Down-Regulation , Dynamin I/genetics , Dynamin I/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Gene Knockdown Techniques , HEK293 Cells , Humans , Pertussis Toxin/administration & dosage , Receptors, Thyrotropin/genetics , Signal Transduction/genetics , beta-Arrestin 2/genetics , beta-Arrestin 2/metabolism
8.
Eur J Immunol ; 49(11): 2030-2043, 2019 11.
Article in English | MEDLINE | ID: mdl-31318439

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are found in the CNS during neuroinflammation and have been reported to exert regulatory functions in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). However, the mechanisms of entry of pDCs into the CNS as well as their phenotype and innate functional properties, once recruited into the CNS, have not been thoroughly examined. Herein, we show that pDCs rapidly accumulate into the brain and spinal cord during the acute phase of EAE, and maintain the expression of numerous phenotypic markers typical of peripheral pDCs. Functionally, CNS-pDCs constitutively expressed IRF7 and were able to rapidly produce type I IFNs and IL-12p40 upon ex vivo TLR-9 stimulation. Using adoptive transfer experiments, we provide evidence that CNS-pDC are recruited from the blood and accumulate into the CNS during the acute phase of EAE. Accumulation of pDCs into the CNS was strongly inhibited in the absence of CD29, but not CD18, suggesting a major role for ß1 but not ß2 integrins. Indeed, blocking the CD49d α4-integrins during acute EAE drastically diminished CNS-pDC numbers. Together, our results demonstrate that circulating pDCs are actively recruited into the CNS during acute EAE through a mechanism largely dependent on CD49d/CD29-integrins.


Subject(s)
Brain/immunology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Integrin alpha4/immunology , Integrin beta1/immunology , Spinal Cord/immunology , Adoptive Transfer , Animals , Brain/drug effects , Brain/pathology , Cell Movement/immunology , Dendritic Cells/drug effects , Dendritic Cells/pathology , Dendritic Cells/transplantation , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression Regulation , Immunity, Innate , Integrin alpha4/genetics , Integrin beta1/genetics , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Interferon Type I/genetics , Interferon Type I/immunology , Interleukin-12 Subunit p40/genetics , Interleukin-12 Subunit p40/immunology , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Peptide Fragments/administration & dosage , Pertussis Toxin/administration & dosage , Signal Transduction , Spinal Cord/drug effects , Spinal Cord/pathology , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology
9.
J Leukoc Biol ; 105(5): 829-841, 2019 05.
Article in English | MEDLINE | ID: mdl-30762897

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are a diverse group of cells that are recognized for their remarkable suppressive effects on pro-inflammatory T cells. The pleiotropic nature of these cells, however, has been demonstrated by their differential effects on immune responses in different settings. Our and others' work has demonstrated suppressive effects of these cells. We previously demonstrated that these cells were mobilized to the lungs during experimental autoimmune encephalomyelitis (EAE), which is a murine model of multiple sclerosis, and potently inhibited CD8+ T cell responses against influenza infection. Interestingly, they appeared to have a lesser effect on CD4+ T cells, and in fact, others have demonstrated that spleen-derived MDSCs could actually promote Th17 differentiation. We sought to determine the role of lung-derived MDSCs on EAE pathogenesis, as excursion through the lungs by pathologic CNS-Ag targeted T cells was shown to be critical for EAE induction. Our results indicate a robust accumulation of granulocytic MDSCs in the lungs of mice during EAE, which could promote Th17 polarization, and which coincided with the trafficking of autoimmune-targeted T cells through the lungs. These studies underscore the pleiotropic effect of MDSCs on T cells and their potential pro-inflammatory phenotypes in neuro-inflammatory disease. Understanding both the intrinsic multifunctional nature of these cells and the ability to influence organ-specific targets such as the CNS from remote organs such as lungs will help to elucidate both mechanisms of disease and possible new therapeutic approaches.


Subject(s)
Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Lung/immunology , Myeloid-Derived Suppressor Cells/immunology , Th17 Cells/immunology , Animals , Autoimmunity , Cell Communication/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Cell Movement , Central Nervous System/pathology , Coculture Techniques , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression , Humans , Interleukin-17/genetics , Interleukin-17/immunology , Lung/pathology , Mice, Inbred C57BL , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/pathology , Peptide Fragments/administration & dosage , Pertussis Toxin/administration & dosage , Signal Transduction/immunology , Th17 Cells/drug effects , Th17 Cells/pathology , Transforming Growth Factor beta/pharmacology
10.
Nat Commun ; 10(1): 229, 2019 01 16.
Article in English | MEDLINE | ID: mdl-30651548

ABSTRACT

There are no conventional lymphatic vessels within the CNS parenchyma, although it has been hypothesized that lymphatics near the cribriform plate or dura maintain fluid homeostasis and immune surveillance during steady-state conditions. However, the role of these lymphatic vessels during neuroinflammation is not well understood. We report that lymphatic vessels near the cribriform plate undergo lymphangiogenesis in a VEGFC - VEGFR3 dependent manner during experimental autoimmune encephalomyelitis (EAE) and drain both CSF and cells that were once in the CNS parenchyma. Lymphangiogenesis also contributes to the drainage of CNS derived antigens that leads to antigen specific T cell proliferation in the draining lymph nodes during EAE. In contrast, meningeal lymphatics do not undergo lymphangiogenesis during EAE, suggesting heterogeneity in CNS lymphatics. We conclude that increased lymphangiogenesis near the cribriform plate can contribute to the management of neuroinflammation-induced fluid accumulation and immune surveillance.


Subject(s)
Brain/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Lymphangiogenesis/immunology , Lymphatic Vessels/immunology , T-Lymphocytes/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antigens/immunology , Antigens/metabolism , Brain/diagnostic imaging , Cell Proliferation , Cerebrospinal Fluid/immunology , Encephalomyelitis, Autoimmune, Experimental/diagnostic imaging , Ethmoid Bone , Evans Blue/administration & dosage , Female , Humans , Immunologic Surveillance/immunology , Lymphatic Vessels/diagnostic imaging , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Myelin-Oligodendrocyte Glycoprotein/immunology , Pertussis Toxin/administration & dosage , Pertussis Toxin/immunology , Vascular Endothelial Growth Factor C/immunology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/immunology , Vascular Endothelial Growth Factor Receptor-3/metabolism
11.
Methods Mol Biol ; 1791: 243-250, 2018.
Article in English | MEDLINE | ID: mdl-30006715

ABSTRACT

Mouse models of peripheral demyelinating neuropathy play an important role in enabling the study of disease pathogenesis. Further, induction in transgenic mice allows for the precise interrogation of disease mechanisms, as well as the analysis of the efficacy and mechanisms of potential new therapies. Here we describe a method to successfully induce experimental autoimmune neuritis (EAN) using myelin protein zero (P0)180-199 peptide in combination with Freund's complete adjuvant and pertussis toxin in the C57BL/6 mouse strain. We also outline a sensitive paradigm of accurately assessing the extent of functional deficits occurring in murine EAN.


Subject(s)
Myelin P0 Protein/immunology , Neuritis, Autoimmune, Experimental/immunology , Peptide Fragments/immunology , Amino Acid Sequence , Animals , Disease Models, Animal , Gait , Mice , Mice, Inbred C57BL , Motor Activity , Myelin P0 Protein/chemistry , Myelin P0 Protein/metabolism , Neuritis, Autoimmune, Experimental/diagnosis , Neuritis, Autoimmune, Experimental/metabolism , Neuritis, Autoimmune, Experimental/physiopathology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Pertussis Toxin/administration & dosage , Pertussis Toxin/adverse effects , Pertussis Toxin/immunology
12.
Cytokine ; 96: 8-15, 2017 08.
Article in English | MEDLINE | ID: mdl-28267649

ABSTRACT

PURPOSE: To evaluate the effect of an intravitreally applied anti-IL-6 antibody for the treatment of experimental autoimmune uveitis (EAU). METHODS: EAU was induced in female B10.RIII mice by Inter-Photoreceptor-Binding-Protein (IRBP) in complete Freund's adjuvant, boosted by Pertussis toxin. Single blinded intravitreal injections of anti-IL-6 antibody were applied 5-7days as well as 8-10days (3day interval) after EAU induction into the randomized treatment eye and phosphate buffered saline (PBS) into the fellow control eye. Clinical and fluorescein angiography scoring (6 EAU grades) was done at each injection day and at enucleation day 14. Enucleated eyes were either scored histologically (6 EAU grades) or examined by ELISA for levels of IL-6, IL-17 and IL-6 soluble Receptor (sIL-6R). RESULTS: Uveitis developed in all 12 mice. Clinical uveitis score was significantly reduced (p=0.035) in treated eyes (median 2.0, range 0-4.0, n=12) compared to the fellow control eyes (median 3.0, range 1.0-4.0, n=12). Angiography scores were reduced in 9/12 treated eyes and histological scores in 3/4 treated eyes compared to the fellow control eyes. Cytokine levels were determined in 8 mice, of which 4 responded to anti-IL-6 treatment and 4 did not respond. All mice responding to treatment had a significant reduction of IL-6 (p<0.01) and IL-17 (p=0.01) levels in treated eyes compared to the fellow control eyes. This difference was not seen in non-responding mice. CONCLUSIONS: Intravitreal anti-IL-6 treatment significantly attenuates experimental autoimmune uveitis in mice. EAU activity correlates with ocular IL-6 and IL-17 levels.


Subject(s)
Antibodies/therapeutic use , Autoimmune Diseases/therapy , Interleukin-6/antagonists & inhibitors , Uveitis/therapy , Animals , Antibodies/administration & dosage , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Eye Proteins/analysis , Eye Proteins/immunology , Female , Freund's Adjuvant , Immunotherapy , Interleukin-17/analysis , Interleukin-17/immunology , Interleukin-6/analysis , Interleukin-6/immunology , Intravitreal Injections , Mice , Pertussis Toxin/administration & dosage , Random Allocation , Uveitis/chemically induced , Uveitis/immunology
13.
Behav Brain Res ; 323: 1-10, 2017 04 14.
Article in English | MEDLINE | ID: mdl-28119127

ABSTRACT

Previous work from our laboratory has shown that nonspecific kappa opioid receptor (KOR) antagonism in primary somatosensory cortex (S1) can inhibit acquisition for the forebrain-dependent associative task, Whisker-Trace Eyeblink conditioning (WTEB). Although studies have demonstrated that KOR activation can alter stimuli salience, our studies controlled for these factors, demonstrating that KOR also plays a role in facilitating learning. KOR has two distinct phases of activation followed by internalization/downregulation, that each independently activate kinases and transcription factors known to mediate task acquisition and memory consolidation respectively. The current study demonstrated that antagonism of the initial phase of KOR activation in S1 via local injections of the g-protein inhibitor, pertussis toxin (PTX), blocked initial WTEB acquisition without affecting retention of the association. In contrast, KOR late phase antagonism in S1 via local injections of the GRK3-specific antagonist, guanidinonaltrindole (GNTI), blocked retention of the WTEB association without affecting task acquisition. Consistent with the known mechanism for KOR activation, KOR protein expression in S1 was found to be decreased following WTEB training, further supporting the involvement of neocortical KOR activation with learning. Prior studies have shown that task acquisition and memory consolidation are mediated by distinct molecular processes; however, little is known regarding a potential mechanism driving these processes. The current study suggests that neocortical KOR activation mediates activation of these processes with learning. This study provides the first evidence for a time- and learning-dependent property of neocortical KOR in facilitating acquisition and consolidation of associative memories, while elucidating an unexplored neocortical learning mechanism.


Subject(s)
Association Learning/physiology , Memory Consolidation/physiology , Receptors, Opioid, kappa/physiology , Somatosensory Cortex/physiology , Animals , Association Learning/drug effects , Conditioning, Classical , Guanidines/administration & dosage , Male , Memory Consolidation/drug effects , Mice, Inbred C57BL , Morphinans/administration & dosage , Pertussis Toxin/administration & dosage , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, kappa/metabolism , Somatosensory Cortex/drug effects , Somatosensory Cortex/metabolism
14.
EMBO J ; 36(1): 102-115, 2017 01 04.
Article in English | MEDLINE | ID: mdl-27827809

ABSTRACT

Interleukin-1 (IL-1) is implicated in numerous pathologies, including multiple sclerosis and its animal model experimental autoimmune encephalomyelitis (EAE). However, the exact mechanism by which IL-1 is involved in the generation of pathogenic T cells and in disease development remains largely unknown. We found that following EAE induction, pertussis toxin administration leads to IL-1 receptor type 1 (IL-1R1)-dependent IL-1ß expression by myeloid cells in the draining lymph nodes. This myeloid-derived IL-1ß did not vitally contribute to the generation and plasticity of Th17 cells, but rather promoted the expansion of a GM-CSF+ Th17 cell subset, thereby enhancing its encephalitogenic potential. Lack of expansion of GM-CSF-producing Th17 cells led to ameliorated disease in mice deficient for IL-1R1 specifically in T cells. Importantly, pathogenicity of IL-1R1-deficient T cells was fully restored by IL-23 polarization and expansion in vitro Therefore, our data demonstrate that IL-1 functions as a mitogenic mediator of encephalitogenic Th17 cells rather than qualitative inducer of their generation.


Subject(s)
Cell Proliferation , Encephalomyelitis, Autoimmune, Experimental/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Interleukin-1/metabolism , Th17 Cells/chemistry , Th17 Cells/physiology , Animals , Mice , Pertussis Toxin/administration & dosage , Pertussis Toxin/toxicity
15.
Nat Commun ; 7: 11904, 2016 06 17.
Article in English | MEDLINE | ID: mdl-27312851

ABSTRACT

Death-associated protein kinase (DAPK) is a tumour suppressor. Here we show that DAPK also inhibits T helper 17 (Th17) and prevents Th17-mediated pathology in a mouse model of autoimmunity. We demonstrate that DAPK specifically downregulates hypoxia-inducible factor 1α (HIF-1α). In contrast to the predominant nuclear localization of HIF-1α in many cell types, HIF-1α is located in both the cytoplasm and nucleus in T cells, allowing for a cytosolic DAPK-HIF-1α interaction. DAPK also binds prolyl hydroxylase domain protein 2 (PHD2) and increases HIF-1α-PHD2 association. DAPK thereby promotes the proline hydroxylation and proteasome degradation of HIF-1α. Consequently, DAPK deficiency leads to excess HIF-1α accumulation, enhanced IL-17 expression and exacerbated experimental autoimmune encephalomyelitis. Additional knockout of HIF-1α restores the normal differentiation of Dapk(-/-) Th17 cells and prevents experimental autoimmune encephalomyelitis development. Our results reveal a mechanism involving DAPK-mediated degradation of cytoplasmic HIF-1α, and suggest that raising DAPK levels could be used for treatment of Th17-associated inflammatory diseases.


Subject(s)
Death-Associated Protein Kinases/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Th17 Cells/immunology , Animals , Death-Associated Protein Kinases/deficiency , Death-Associated Protein Kinases/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Hydroxylation , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Hypoxia-Inducible Factor-Proline Dioxygenases/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Jurkat Cells , Mice , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Peptide Fragments/administration & dosage , Pertussis Toxin/administration & dosage , Proline/metabolism , Proteasome Endopeptidase Complex , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th17 Cells/drug effects , Th17 Cells/pathology
16.
Neurosci Res ; 89: 31-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25218563

ABSTRACT

The effect of cholera toxin (CTX) or pertussis toxin (PTX) administered supraspinally on hippocampal neuronal cell death in CA3 region induced by kainic acid (KA) was examined in mice. After the pretreatment with either PTX or CTX intracerebroventricularly (i.c.v.), mice were administered i.c.v. with KA. The i.c.v. treatment with KA caused a neuronal cell death in CA3 region and PTX, but not CTX, attenuated the KA-induced neuronal cell death. In addition, i.c.v. treatment with KA caused an elevation of the blood glucose level. The i.c.v. PTX pretreatment alone caused a hypoglycemia and inhibited KA-induced hyperglycemic effect. However, i.c.v. pretreatment with CTX did not affect the basal blood glucose level and KA-induced hyperglycemic effect. Moreover, KA administered i.c.v. caused an elevation of corticosterone level and reduction of the blood insulin level. Whereas, i.c.v. pretreatment with PTX further enhanced KA-induced up-regulation of corticosterone level. Furthermore, i.c.v. administration of PTX alone increased the insulin level and KA-induced hypoinsulinemic effect was reversed. In addition, PTX pretreatment reduces the KA-induced seizure activity. Our results suggest that supraspinally administered PTX, exerts neuroprotective effect against KA-induced neuronal cells death in CA3 region and neuroprotective effect of PTX is mediated by the reduction of KA-induced blood glucose level.


Subject(s)
CA3 Region, Hippocampal/drug effects , Cell Death/drug effects , Cholera Toxin/administration & dosage , Kainic Acid/toxicity , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Pertussis Toxin/administration & dosage , Animals , Blood Glucose/drug effects , Corticosterone/blood , Hyperglycemia/chemically induced , Hypoglycemia/chemically induced , Infusions, Intraventricular , Insulin/blood , Male , Mice , Mice, Inbred ICR , Seizures/chemically induced
17.
Pharmacology ; 94(1-2): 29-40, 2014.
Article in English | MEDLINE | ID: mdl-25171426

ABSTRACT

BACKGROUND/AIMS: To show whether intrathecal (i.t.) treatment with pertussis toxin (PTX) produces a hypoglycemic effect in ICR, db/db and streptozotocin-treated mice. METHODS: The blood glucose level (BGL) was measured after i.t. treatment with PTX, AB5 toxins and PTX subunits. Insulin or leptin levels were measured after PTX injection. The effect of PTX on the BGL was examined in adrenalectomized (ADX) mice. Glucose transporter (GLUT) levels were determined by Western blotting. RESULTS: PTX attenuated the elevated BGL in the D-glucose-fed model in a long-term manner. Heat-labile toxin (HLT), HLT subunit B or Shiga toxin, which belong to the AB5 toxins, administered i.t. did not affect the BGL. PTX A protomer (PTX-A) or PTX B oligomers (PTX-B) injected i.t. did not have an effect on the BGL as well. However, combined treatment with PTX-A and PTX-B subunits caused a hypoglycemic effect. The leptin level was gradually reduced by PTX for up to 6 days, without affecting the insulin level. PTX administered i.t. significantly decreased the BGL further in ADX mice. Moreover, GLUT-2 (hypothalamus and pituitary gland), GLUT-4 (muscle) and GLUT-3 (adrenal gland) expression levels were increased, whereas GLUT-1 (brain cortex, liver, muscle and spinal cord), GLUT-2 (liver) and GLUT-3 (brain cortex and pituitary gland) expression levels were decreased. DISCUSSION: Our data suggest that PTX administered spinally produces a hypoglycemic effect in a long-term manner, and PTX-induced hypoglycemia appears to be mediated by the reduction in activity of the glucocorticoid system. Furthermore, PTX may modulate the insulin level during hypoglycemia. Among GLUTs, GLUT-4 in muscle, GLUT-2 in the liver, hypothalamus and pituitary gland as well as GLUT-1 in the adrenal gland may be responsible for PTX-induced hypoglycemia.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Hypoglycemia/chemically induced , Hypoglycemic Agents/pharmacology , Pertussis Toxin/pharmacology , Animals , Blood Glucose/drug effects , Blotting, Western , Glucose Transport Proteins, Facilitative/metabolism , Hypoglycemic Agents/administration & dosage , Injections, Spinal , Insulin/metabolism , Leptin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Pertussis Toxin/administration & dosage , Streptozocin , Time Factors
18.
FASEB J ; 28(4): 1898-909, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24371118

ABSTRACT

Histamine and its receptors are important in both multiple sclerosis and experimental allergic encephalomyelitis (EAE). C57BL/6J (B6) mice deficient for the histamine H2 receptor (H2RKO) are less susceptible to EAE and exhibit blunted Th1 responses. However, whether decreased antigen-specific T-cell effector responses in H2RKO mice were due to a lack of H2R signaling in CD4(+) T cells or antigen-presenting cells has remained unclear. We generated transgenic mice expressing H2R specifically in T cells on the H2RKO background, and, using wild-type B6 and H2RKO mice as controls, induced EAE either in the presence or absence of the ancillary adjuvant pertussis toxin (PTX), which models the effects of infectious inflammatory stimuli on autoimmune disease. We monitored the mice for clinical signs of EAE and neuropathology, as well as effector T-cell responses using flow cytometry. EAE severity and neuropathology in H2RKO mice expressing H2R exclusively in T cells become equal to those in wild-type B6 mice only when PTX is used to elicit disease. EAE complementation was associated with frequencies of CD4(+)IFN-γ(+) and CD4(+)IL-17(+) cells that are equal to or greater than those in wild-type B6, respectively. Thus, the regulation of encephalitogenic T-cell responses and EAE susceptibility by H2R signaling in CD4(+) T cells is dependent on gene × environment interactions.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Receptors, Histamine H2/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Flow Cytometry , Gene Expression/immunology , Genetic Predisposition to Disease , HEK293 Cells , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Myelin-Oligodendrocyte Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein/toxicity , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Peptide Fragments/toxicity , Pertussis Toxin/administration & dosage , Pertussis Toxin/immunology , Receptors, Histamine H2/genetics , Receptors, Histamine H2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Severity of Illness Index , Signal Transduction/genetics , T-Lymphocytes/metabolism
19.
PLoS One ; 8(12): e83457, 2013.
Article in English | MEDLINE | ID: mdl-24386204

ABSTRACT

Epstein-Barr virus induced receptor 2 (EBI2), a Gαi-coupled G protein-coupled receptor, is a chemotactic receptor for B, T and dendritic cells (DC). Genetic studies have also implicated EBI2 as a regulator of an interferon regulatory factor 7 (IRF7)-driven inflammatory network (IDIN) associated with autoimmune diseases, although the corollary in primary type I IFN-producing cells has not been reported. Here we demonstrate that EBI2 negatively regulates type I IFN responses in plasmacytoid DC (pDCs) and CD11b(+) myeloid cells. Activation of EBI2(-/-) pDCs and CD11b(+) cells with various TLR ligands induced elevated type I IFN production compared to wild-type cells. Moreover, in vivo challenge with endosomal TLR agonists or infection with lymphocytic choriomeningitis virus elicited more type I IFNs and proinflammatory cytokines in EBI2(-/-) mice compared to normal mice. Elevated systemic cytokines occurred despite impaired ability of EBI2-deficient pDCs and CD11b(+) cells to migrate from the blood to the spleen and peritoneal cavity under homeostatic conditions. As reported for other immune cells, pDC migration was dependent on the ligand for EBI2, 7α,25-dihydroxycholesterol. Consistent with a cell intrinsic role for EBI2, type I IFN-producing cells from EBI2-deficient mice expressed higher levels of IRF7 and IDIN genes. Together these data suggest a negative regulatory role for EBI2 in balancing TLR-mediated responses to foreign and to self nucleic acids that may precipitate autoimmunity.


Subject(s)
Dendritic Cells/metabolism , Interferon Type I/metabolism , Myeloid Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Chemotaxis/drug effects , Chemotaxis/genetics , Chemotaxis/immunology , Dendritic Cells/immunology , Female , Gene Expression , Interferon Regulatory Factor-7/metabolism , Ligands , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/metabolism , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Knockout , Myeloid Cells/immunology , Peritoneal Cavity , Pertussis Toxin/administration & dosage , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Spleen/immunology , Spleen/metabolism , Toll-Like Receptors/metabolism
20.
J Immunol ; 189(6): 2702-6, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22896631

ABSTRACT

Nonlymphoid T cell populations control local infections and contribute to inflammatory diseases, thus driving efforts to understand the regulation of their migration, differentiation, and maintenance. Numerous observations indicate that T cell trafficking and differentiation within the lung are starkly different from what has been described in most nonlymphoid tissues, including intestine and skin. After systemic infection, we found that >95% of memory CD8 T cells isolated from mouse lung via standard methods were actually confined to the pulmonary vasculature, despite perfusion. A respiratory route of challenge increased virus-specific T cell localization within lung tissue, although only transiently. Removing blood-borne cells from analysis by the simple technique of intravascular staining revealed distinct phenotypic signatures and chemokine-dependent trafficking restricted to Ag-experienced T cells. These results precipitate a revised model for pulmonary T cell trafficking and differentiation and a re-evaluation of studies examining the contributions of pulmonary T cells to protection and disease.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Capillaries/immunology , Capillaries/pathology , Lung/blood supply , Lung/immunology , Animals , Antibodies/administration & dosage , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/transplantation , Capillaries/virology , Cell Movement/genetics , Cell Movement/immunology , Lung/metabolism , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Pertussis Toxin/administration & dosage , Pneumonia, Viral/genetics , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology , Spleen/immunology , Spleen/pathology , Spleen/transplantation , Staining and Labeling/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...