Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Int Immunopharmacol ; 101(Pt A): 108280, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34710845

ABSTRACT

The COVID-19 pandemic, caused by a highly virulent and transmissible pathogen, has proven to be devastating to society. Mucosal vaccines that can induce antigen-specific immune responses in both the systemic and mucosal compartments are considered an effective measure to overcome infectious diseases caused by pathogenic microbes. We have recently developed a nasal vaccine system using cationic liposomes composed of 1,2-dioleoyl-3-trimethylammonium-propane and cholesteryl 3ß-N-(dimethylaminoethyl)carbamate in mice. However, the comprehensive molecular mechanism(s), especially the host soluble mediator involved in this process, by which cationic liposomes promote antigen-specific mucosal immune responses, remain to be elucidated. Herein, we show that intranasal administration of cationic liposomes elicited interleukin-6 (IL-6) expression at the site of administration. Additionally, both nasal passages and splenocytes from mice nasally immunized with cationic liposomes plus ovalbumin (OVA) were polarized to produce IL-6 when re-stimulated with OVA in vitro. Furthermore, pretreatment with anti-IL-6R antibody, which blocks the biological activities of IL-6, attenuated the production of OVA-specific nasal immunoglobulin A (IgA) but not OVA-specific serum immunoglobulin G (IgG) responses. In this study, we demonstrated that IL-6, exerted by nasally administered cationic liposomes, plays a crucial role in antigen-specific IgA induction.


Subject(s)
Immunity, Mucosal/immunology , Immunoglobulin A/metabolism , Interleukin-6/immunology , Vaccines/immunology , Administration, Intranasal , Animals , Antibody Formation/drug effects , Antigens/immunology , COVID-19/prevention & control , Cations/immunology , Cations/therapeutic use , Fatty Acids, Monounsaturated/immunology , Fatty Acids, Monounsaturated/therapeutic use , Female , Immunity, Mucosal/drug effects , Immunoglobulin G/blood , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Interleukin-6/metabolism , Liposomes/immunology , Liposomes/therapeutic use , Mice , Nasal Mucosa/immunology , Nasal Mucosa/metabolism , Ovalbumin/immunology , Quaternary Ammonium Compounds/immunology , Quaternary Ammonium Compounds/therapeutic use , Spleen/metabolism , Vaccines/administration & dosage
2.
Cell Rep ; 34(1): 108601, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33406440

ABSTRACT

Immune cells can metabolize glucose, amino acids, and fatty acids (FAs) to generate energy. The roles of different FA species and their impacts on humoral immunity remain poorly understood. Here, we report that proliferating B cells require monounsaturated FAs (MUFAs) to maintain mitochondrial metabolism and mTOR activity and to prevent excessive autophagy and endoplasmic reticulum (ER) stress. Furthermore, B cell-extrinsic stearoyl-CoA desaturase (SCD) activity generates MUFA to support early B cell development and germinal center (GC) formation in vivo during immunization and influenza infection. Thus, SCD-mediated MUFA production is critical for humoral immunity.


Subject(s)
B-Lymphocytes/physiology , Fatty Acids, Monounsaturated/immunology , Fatty Acids, Monounsaturated/metabolism , Immunity, Humoral , Mitochondria/physiology , Stearoyl-CoA Desaturase/physiology , TOR Serine-Threonine Kinases/metabolism , Animals , Autophagy , Endoplasmic Reticulum Stress , Gene Knockout Techniques , Mice , Mice, Inbred C57BL , Mice, Transgenic , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/immunology , TOR Serine-Threonine Kinases/immunology
4.
Int Immunopharmacol ; 84: 106552, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32422526

ABSTRACT

PURPOSE: To study the role of palmitoleic acid (PA) in the pathogenesis of acute anterior uveitis (AAU). METHODS: PA levels in feces from AAU patients were measured by gas chromatography coupled with a mass spectrometer (GC-MS) and compared with samples obtained from healthy individuals. Enzyme linked immunosorbent assay (ELISA) and flow cytometry (FCM) were used to assess the effect of PA on dendritic cells (DCs) and CD4+T cells obtained from mice, AAU patients and healthy individuals. C57BL/6 mice were fed with PA or vehicle and experimental autoimmune uveitis (EAU) was induced with a human retinal IRBP651-670 peptide. Disease severity of EAU was evaluated by clinical manifestation and histology. Differentiation of splenic Type 1 helper T cells (Th1) and Th17 cells was evaluated by FCM. Tandem mass tag (TMT)-based proteomics analysis was used to identify differentially expressed proteins following incubation of DCs with PA. RESULTS: The fecal concentration of PA was increased in AAU patients as compared with healthy individuals. In vitro, PA promoted apoptosis of DCs and inhibited the secretion of TNF-α from mouse bone-marrow-derived dendritic cells (BMDCs) as well as in DCs from AAU patients and healthy individuals. It only decreased DCs surface marker expression and IL-12p70 secretion in BMDCs and healthy individuals DCs but not in AAU patient DCs. PA-treated BMDCs inhibited Th cell differentiation from mouse naïve CD4+T cells and IL-17 and IFN-γ secretion in co-culture supernatants. PA also inhibited the differentiation of Th cells and secretion of IFN-γ and IL-17 in CD4+T cells from mice, AAU patients and healthy individuals. In vivo, PA-treated EAU mice showed milder clinical and histopathological intraocular manifestations as compared with the control group. PA feeding inhibited differentiation of splenic Th17 cells, whereas Th1 cells were not affected. Up to 30 upregulated and 77 downregulated proteins were identified when comparing PA-treated DCs with controls. CONCLUSION: An increased expression of fecal PA was observed in AAU patients. PA was shown to have immunoregulatory effects on DCs and CD4+T cells and attenuated disease severity in EAU mice.


Subject(s)
Fatty Acids, Monounsaturated/immunology , Feces/chemistry , Immunologic Factors/immunology , Uveitis, Anterior/immunology , Adult , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Dendritic Cells/immunology , Female , Humans , Male , Mice, Inbred C57BL , Young Adult
5.
Dev Comp Immunol ; 107: 103643, 2020 06.
Article in English | MEDLINE | ID: mdl-32067998

ABSTRACT

Eicosanoids mediate both cellular and humoral immune responses in insects. Epoxyeicosatrienoic acids (EETs) are a group of eicosanoids containing epoxide formed by epoxygenase (EPX) activity of cytochrome P450 (CYP). Although EETs have been considered to mediate immune responses in some insects, their synthetic machinery was little understood in insects. This study monitored EETs in a lepidopteran insect, Spodoptera exigua, immunized with bacteria and found all four EETs (5,6-EET, 8,9-EET, 11,12-EET, and 14,15-EET) from larval fat body at 247-1,736 pg/g levels. Then to predict EPXs, 140 CYPs were collected from S. exigua transcriptomes and compared with human EPXs. Four CYPs (SeEPX1-SeEPX4) sharing homologies with human EPXs were chosen and assessed in subsequent expression and functional analyses. All four EPXs were expressed in all development stages. In larval stage, all four EPXs were expressed in immune-associated tissues such as fat body and hemocytes. Furthermore, their expression levels were highly enhanced by bacterial challenge in different tissues. RNA interference (RNAi) using gene-specific double stranded RNA injection suppressed their expression levels by more than 55%. RNAi treatments interfered with hemocyte-spreading behavior and nodule formation upon bacterial challenge except RNAi treatment against SeEPX2. All four EETs stimulated cellular immune response measured by nodule formation in S. exigua. The suppressed immune responses by the RNAi treatments against three SeEPXs were rescued by the addition of 8,9-EET. However, other three EETs gave their specific rescue effect depending on SeEPX types under RNAi. In humoral immune response, all four RNAi treatments suppressed expression of antimicrobial peptide genes. This study reports the presence of all four EETs in larval fat body of S. exigua and suggests that four SeEPXs are associated with immune responses mediated by EETs.


Subject(s)
Epoxy Compounds/metabolism , Fat Body/metabolism , Fatty Acids, Monounsaturated/metabolism , Hemocytes/metabolism , Spodoptera/immunology , Animals , Epoxy Compounds/immunology , Fatty Acids, Monounsaturated/immunology , Humans , Immunity, Innate , Insect Proteins/genetics , Insect Proteins/metabolism , Larva , Oxidoreductases/genetics , Oxidoreductases/metabolism , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , RNA, Small Interfering/genetics , Transcriptome
6.
J Autoimmun ; 91: 83-96, 2018 07.
Article in English | MEDLINE | ID: mdl-29753567

ABSTRACT

Fibrosis remains a serious health concern in patients with chronic liver disease. We recently reported that chemically induced chronic murine liver injury triggers increased expression of junctional adhesion molecules (JAMs) JAM-B and JAM-C by endothelial cells and de novo synthesis of JAM-C by hepatic stellate cells (HSCs). Here, we demonstrate that biopsies of patients suffering from primary biliary cholangitis (PBC), primary sclerosing cholangitis (PSC) or autoimmune hepatitis (AIH) display elevated levels of JAM-C on portal fibroblasts (PFs), HSCs, endothelial cells and cholangiocytes, whereas smooth muscle cells expressed JAM-C constitutively. Therefore, localization and function of JAM-B and JAM-C were investigated in three mouse models of autoimmune-driven liver inflammation. A PBC-like disease was induced by immunization with 2-octynoic acid-BSA conjugate, which resulted in the upregulation of both JAMs in fibrotic portal triads. Analysis of a murine model of PSC revealed a role of JAM-C in PF cell-cell adhesion and contractility. In mice suffering from AIH, endothelial cells increased JAM-B level and HSCs and capsular fibroblasts became JAM-C-positive. Most importantly, AIH-mediated liver fibrosis was reduced in JAM-B-/- mice or when JAM-C was blocked by soluble recombinant JAM-C. Interestingly, loss of JAM-B/JAM-C function had no effect on leukocyte infiltration, suggesting that the well-documented function of JAMs in leukocyte recruitment to inflamed tissue was not effective in the tested chronic models. This might be different in patients and may even be complicated by the fact that human leukocytes express JAM-C. Our findings delineate JAM-C as a mediator of myofibroblast-operated contraction of the liver capsule, intrahepatic vasoconstriction and bile duct stricture. Due to its potential to interact heterophilically with endothelial JAM-B, JAM-C supports also HSC/PF mural cell function. Together, these properties allow JAM-B and JAM-C to actively participate in vascular remodeling associated with liver/biliary fibrosis and suggest them as valuable targets for anti-fibrosis therapies.


Subject(s)
Cell Adhesion Molecules/metabolism , Cholangitis, Sclerosing/metabolism , Endothelial Cells/metabolism , Hepatitis, Autoimmune/metabolism , Immunoglobulins/metabolism , Inflammation/metabolism , Liver Cirrhosis, Biliary/metabolism , Liver/pathology , Myocytes, Smooth Muscle/metabolism , Myofibroblasts/metabolism , Animals , Cell Adhesion , Cell Adhesion Molecules/genetics , Cells, Cultured , Disease Models, Animal , Fatty Acids, Monounsaturated/immunology , Female , Fibrosis , Humans , Immunoglobulins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Vascular Remodeling , Vasoconstriction
7.
Parasite Immunol ; 39(11)2017 Nov.
Article in English | MEDLINE | ID: mdl-28921566

ABSTRACT

Although there have been several attempts to develop a vaccine against leishmaniasis, no vaccine in human has been developed yet. Liposomes consisting of 1, 2-dioleoyl-3-trimethylammonium-propane (DOTAP) encapsulating soluble Leishmania antigens (SLA) enhance protective immunity of SLA against Leishmania major infection in mice. However, they immobilized at the injection site because of their positive charge. To overcome the problem, shielding the surface charge with polyethylene glycol (PEGylation) was chosen in this study. Liposomal SLA consisting different concentrations of PEG (1.9%-15% mol) were prepared. BALB/c mice were immunized three times in 3 weeks intervals with different formulations. Lesion development and parasite burden in footpad and spleen were evaluated to specify the type of generated immune response and extent of protection. Th1/Th2 cytokine profiles and IgG isotypes were also analysed. The maximum protection was observed in mice immunized with Lip-SLA or pLip-SLA (1.9%) due to smaller footpad swelling, reduction in parasite load, an increase in IgG2a and IFN-γ production. Our results showed that immunization of mice with a high level of PEG (>7.5%) did not improve protective immunity of liposomal SLA. The presence of PEG, particularly more than 3.75%, is not recommended for protection against leishmaniasis.


Subject(s)
Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Immunoglobulin G/blood , Leishmania major/immunology , Leishmaniasis Vaccines/immunology , Liposomes/immunology , Polyethylene Glycols/chemistry , Adjuvants, Immunologic/metabolism , Animals , Antibodies, Protozoan/immunology , Cytokines/blood , Cytokines/immunology , Disease Models, Animal , Fatty Acids, Monounsaturated/chemistry , Fatty Acids, Monounsaturated/immunology , Female , Immunoglobulin G/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Leishmaniasis/immunology , Leishmaniasis/parasitology , Leishmaniasis/prevention & control , Liposomes/chemistry , Mice , Mice, Inbred BALB C , Parasite Load , Quaternary Ammonium Compounds/chemistry , Quaternary Ammonium Compounds/immunology , Spleen/cytology , Spleen/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Vaccination
8.
Eur J Immunol ; 47(11): 1890-1899, 2017 11.
Article in English | MEDLINE | ID: mdl-28762497

ABSTRACT

The development of vaccines for infectious diseases for which we currently have none, including HIV, will likely require the use of adjuvants that strongly promote germinal center responses and somatic hypermutation to produce broadly neutralizing antibodies. Here we compared the outcome of immunization with the T-cell dependent antigen, NP-conjugated to chicken gamma globulin (NP-CGG) adjuvanted with the toll-like receptor 9 (TLR9) ligands, CpG-A or CpG-B, alone or conjugated with the cationic lipid carrier, DOTAP. We provide evidence that only NP-CGG adjuvanted with DOTAP-CpG-B was an effective vaccine in mice resulting in robust germinal center responses, isotype switching and high affinity NP-specific antibodies. The effectiveness of DOTAP-CpG-B as an adjuvant was dependent on the expression of the TLR9 signaling adaptor MyD88 in immunized mice. These results indicate DOTAP-CpG-B but not DOTAP-CpG-A is an effective adjuvant for T cell-dependent protein antigen-based vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Fatty Acids, Monounsaturated/pharmacology , Oligodeoxyribonucleotides/immunology , Quaternary Ammonium Compounds/pharmacology , T-Lymphocytes/immunology , Vaccines/immunology , Animals , Antibody Affinity , Fatty Acids, Monounsaturated/immunology , Germinal Center/immunology , Mice , Mice, Inbred C57BL , Oligodeoxyribonucleotides/pharmacology , Quaternary Ammonium Compounds/immunology , Vaccines/pharmacology
9.
Muscle Nerve ; 55(4): 532-538, 2017 04.
Article in English | MEDLINE | ID: mdl-27511866

ABSTRACT

INTRODUCTION: The diagnostic sensitivity of repetitive nerve stimulation (RNS) in patients with myasthenia gravis (MG) varies as a function of the number of muscles or the choice of muscles studied. METHODS: By exploring 12 muscles bilaterally, we evaluated the global sensitivity of RNS at rest, the sensitivity in different clinical forms, and the sensitivity of different combinations of muscles studied. RESULTS: The global sensitivity of RNS was 82%, and specificity was 100%. The sensitivity in the MG subgroups was as follows: ocular (O) = 67%; oculobulbar (OB) = 86%; and generalized (G) = 89%. The most sensitive muscles were the anconeus in group O, orbicularis oculi (OO) or nasalis in group OB, and the trapezius in group G. Maximum sensitivity was obtained by exploring OO, trapezius, and anconeus bilaterally. CONCLUSIONS: We recommend bilateral exploration of at least 3 muscles, a facial muscle, trapezius, and anconeus. Muscle Nerve 55: 532-538, 2017.


Subject(s)
Electric Stimulation/methods , Muscle, Skeletal/physiopathology , Myasthenia Gravis/diagnosis , Adolescent , Adult , Aged , Aged, 80 and over , Autoantibodies/blood , Electromyography , Fatty Acids, Monounsaturated/immunology , Female , Humans , Male , Middle Aged , Neurologic Examination , Receptors, Cholinergic/immunology , Retrospective Studies , Sensitivity and Specificity , Severity of Illness Index , Young Adult
10.
Sci Rep ; 6: 34884, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27721424

ABSTRACT

Primary biliary cirrhosis (PBC) is a progressive autoimmune liver disease with a long natural history. The pathogenesis of PBC is thought to be orchestrated by Th1 and/or Th17. In this study, we investigated the role of CD4+ helper T subsets and their cytokines on PBC using our previous established murine model of 2-OA-OVA immunization. We prepared adeno-associated virus (AAV)-IFN-γ and AAV-IL-4 and studied their individual influences on the natural history of autoimmune cholangitis in this model. Administration of IFN-γ significantly promotes recruitment and lymphocyte activation in the earliest phases of autoimmune cholangitis but subsequently leads to downregulation of chronic inflammation through induction of the immunosuppressive molecule IL-30. In contrast, the administration of IL-4 does not alter the initiation of autoimmune cholangitis, but does contribute to the exacerbation of chronic liver inflammation and fibrosis. Thus Th1 cells and IFN-γ are the dominant contributors in the initiation phase of this model but clearly may have different effects as the disease progress. In conclusion, better understanding of the mechanisms by which helper T cells function in the natural history of cholangitis is essential and illustrates that precision medicine may be needed for patients with PBC at various stages of their disease process.


Subject(s)
Autoimmune Diseases/etiology , Cholangitis/etiology , Interferon-gamma/metabolism , Interleukin-4/metabolism , Interleukins/metabolism , Animals , Cholangitis/immunology , Dependovirus/genetics , Disease Models, Animal , Fatty Acids, Monounsaturated/immunology , Female , Fibrosis/drug therapy , Hepatitis/drug therapy , Interferon-gamma/genetics , Interferon-gamma/pharmacology , Interleukin-4/genetics , Interleukin-4/pharmacology , Interleukins/immunology , Liver/immunology , Liver/metabolism , Liver/pathology , Mice, Inbred C57BL , Ovalbumin/immunology , Precision Medicine , Th1 Cells/immunology , Th1 Cells/metabolism
11.
Sci Rep ; 6: 23348, 2016 Mar 21.
Article in English | MEDLINE | ID: mdl-26996208

ABSTRACT

Galectin-3 (Gal-3) is a carbohydrate binding lectin, with multiple roles in inflammatory diseases and autoimmunity including its antiapoptotic effect on epithelial cells. In particular, increased expression of Gal-3 in epithelial cells is protective from apoptosis. Based on the thesis that apoptosis of biliary epithelial cells (BECs) is critical to the pathogenesis of Primary Biliary Cholangitis (PBC), we have analyzed the role of Gal-3 in the murine model of autoimmune cholangitis. We took advantage of Gal-3 knockout mice and immunized them with a mimotope of the major mitochondrial autoantigen of PBC, 2-octynoic acid (2-OA) coupled to BSA (2OA-BSA) and evaluated the natural history of subsequent disease, compared to control wild-type mice, by measuring levels of antibodies to PDC-E2, immunohistology of liver, and expression of Gal-3. We report herein that deletion of Gal-3 significantly exacerbates autoimmune cholangitis in these mice. This is manifested by increased periportal infiltrations, bile duct damage, granulomas and fibrosis. Interestingly, the BECs of Gal-3 knockout mice had a higher response to apoptotic stimuli and there were more pro-inflammatory lymphocytes and dendritic cells (DCs) in the livers of Gal-3 knockout mice. In conclusion, Gal-3 plays a protective role in the pathways that lead to the inflammatory destruction of biliary epithelial cells.


Subject(s)
Apoptosis , Cholangitis, Sclerosing/immunology , Epithelial Cells/immunology , Galectin 3/immunology , Animals , Autoantigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cholangitis, Sclerosing/pathology , Dihydrolipoyllysine-Residue Acetyltransferase/immunology , Disease Models, Animal , Epithelial Cells/metabolism , Fatty Acids, Monounsaturated/administration & dosage , Fatty Acids, Monounsaturated/immunology , Female , Galectin 3/genetics , Interleukin-13/blood , Interleukin-17/blood , Liver/immunology , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Xenobiotics/immunology
12.
J Gen Virol ; 97(4): 934-940, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26864442

ABSTRACT

Activation of CD8(+)T-cells is an essential part of immune responses elicited by recombinant modified vaccinia virus Ankara (MVA). Strategies to enhance T-cell responses to antigens may be particularly necessary for broadly protective immunization against influenza A virus infections or for candidate vaccines targeting chronic infections and cancer. Here, we tested recombinant MVAs that targeted a model antigen, GFP, to different localizations in infected cells. In vitro characterization demonstrated that GFP accumulated in the nucleus (MVA-nls-GFP), associated with cellular membranes (MVA-myr-GFP) or was equally distributed throughout the cell (MVA-GFP). On vaccination, we found significantly higher levels of GFP-specific CD8(+)T-cells in MVA-myr-GFP-vaccinated BALB/c mice than in those immunized with MVA-GFP or MVA-nls-GFP. Thus, myristoyl modification may be a useful strategy to enhance CD8(+)T-cell responses to MVA-delivered target antigens.


Subject(s)
Antigens/chemistry , CD8-Positive T-Lymphocytes/immunology , Green Fluorescent Proteins/immunology , Protein Processing, Post-Translational/immunology , Vaccinia virus/genetics , Viral Vaccines/immunology , Animals , Antigens/genetics , Antigens/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/ultrastructure , CD8-Positive T-Lymphocytes/virology , Cell Line , Cell Nucleus/immunology , Cell Nucleus/ultrastructure , Chick Embryo , Fatty Acids, Monounsaturated/immunology , Fatty Acids, Monounsaturated/metabolism , Female , Fibroblasts/immunology , Fibroblasts/virology , Green Fluorescent Proteins/administration & dosage , Green Fluorescent Proteins/chemistry , Immunity, Cellular/drug effects , Lymphocyte Count , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Vaccination , Vaccines, Synthetic , Vaccinia virus/immunology , Viral Vaccines/administration & dosage
13.
Iran J Immunol ; 12(4): 274-87, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26714419

ABSTRACT

BACKGROUND: Cationic immune stimulating complexes (PLUSCOMs) are particulate antigen delivery systems. PLUSCOMs consist of cationic immunostimulatory complexes (ISCOMs) derivatives and are able to elicit in vivo T cell responses against an antigen. OBJECTIVE: To evaluate the effects of PLUSCOMs containing Leishmania major antigens (SLA) on the type of immune response generated in the murine model of leishmaniasis. METHODS: PLUSCOMs consisting of 1, 2-dioleoyl-3-trimethylammonium-propane (DOTAP) were used as antigen delivery system/immunoadjuvants for soluble SLA. BALB/c mice were immunized subcutaneously, three times in 2-week intervals. Footpads swellings at the site of challenge and parasite loads were assessed as a measure of protection. The immune responses were also evaluated by determination of IgG subclasses and the level of IFN-γ and IL-4 in cultured splenocytes. RESULTS: There was no significant difference (p<0.05) between the sizes of lesions in mice immunized with different formulations. Also, there was no significant difference in the number of parasites in the footpad or spleen of all groups compared with the control group. The highest level of IFN-γ secretion was observed in the splenocytes of mice immunized with PLUSCOM/SLA (p<0.001) and lower amounts of IL-4 was observed in PLUSCOM group (p<0.001) as compared to negative control. CONCLUSION: Our results indicated that SLA in different formulations generated an immune response with mixed Th1/Th2 response that was not protective enough despite the activation of CD4+ T cells with secreting IFN-γ in groups which received PLUSCOM with antigen.


Subject(s)
Antigens, Protozoan/metabolism , CD4-Positive T-Lymphocytes/drug effects , Fatty Acids, Monounsaturated/metabolism , Immunotherapy , Leishmania/immunology , Leishmaniasis/therapy , Multiprotein Complexes/metabolism , Quaternary Ammonium Compounds/metabolism , Adjuvants, Immunologic , Animals , Antigens, Protozoan/immunology , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Disease Models, Animal , Drug Delivery Systems , Fatty Acids, Monounsaturated/immunology , Female , Immunization , Immunoglobulin G/blood , Interferon-gamma/metabolism , Interleukin-4/metabolism , Leishmaniasis/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Multiprotein Complexes/immunology , Parasite Load , Quaternary Ammonium Compounds/immunology
14.
PLoS One ; 10(10): e0139785, 2015.
Article in English | MEDLINE | ID: mdl-26440657

ABSTRACT

Despite the progress made by modern medicine, infectious diseases remain one of the most important threats to human health. Vaccination against pathogens is one of the primary methods used to prevent and treat infectious diseases that cause illness and death. Vaccines administered by the mucosal route are potentially a promising strategy to combat infectious diseases since mucosal surfaces are a major route of entry for most pathogens. However, this route of vaccination is not widely used in the clinic due to the lack of a safe and effective mucosal adjuvant. Therefore, the development of safe and effective mucosal adjuvants is key to preventing infectious diseases by enabling the use of mucosal vaccines in the clinic. In this study, we show that intranasal administration of a cationic liposome composed of 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) and 3ß-[N-(N',N'-dimethylaminoethane)-carbamoyl] (DC-chol) (DOTAP/DC-chol liposome) has a potent mucosal adjuvant effect in mice. Intranasal vaccination with ovalbumin (OVA) in combination with DOTAP/DC-chol liposomes induced the production of OVA-specific IgA in nasal tissues and increased serum IgG1 levels, suggesting that the cationic DOTAP/DC-chol liposome leads to the induction of a Th2 immune response. Additionally, nasal-associated lymphoid tissue and splenocytes from mice treated with OVA plus DOTAP/DC-chol liposome showed high levels of IL-4 expression. DOTAP/DC-chol liposomes also enhanced OVA uptake by CD11c+ dendritic cells in nasal-associated lymphoid tissue. These data demonstrate that DOTAP/DC-chol liposomes elicit immune responses via an antigen-specific Th2 reaction. These results suggest that cationic liposomes merit further development as a mucosal adjuvant for vaccination against infectious diseases.


Subject(s)
Cholesterol/analogs & derivatives , Fatty Acids, Monounsaturated/immunology , Immunity, Active/immunology , Liposomes/immunology , Ovalbumin/immunology , Quaternary Ammonium Compounds/immunology , Vaccination , Vaccines/immunology , Adjuvants, Immunologic , Administration, Intranasal , Animals , Cholesterol/immunology , Interleukin-4/metabolism , Mice , Th2 Cells/immunology
15.
J Biol Chem ; 290(27): 16979-88, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-25987561

ABSTRACT

A rise in tissue-embedded macrophages displaying "M1-like" proinflammatory polarization is a hallmark of metabolic inflammation during a high fat diet or obesity. Here we show that bone marrow-derived macrophages (BMDM) from high fat-fed mice retain a memory of their dietary environment in vivo (displaying the elevated proinflammatory genes Cxcl1, Il6, Tnf, Nos2) despite 7-day differentiation and proliferation ex vivo. Notably, 6-h incubation with palmitoleate (PO) reversed the proinflammatory gene expression and cytokine secretion seen in BMDM from high fat-fed mice. BMDM from low fat-fed mice exposed to palmitate (PA) for 18 h ex vivo also showed elevated expression of proinflammatory genes (Cxcl1, Il6, Tnf, Nos2, and Il12b) associated with M1 polarization. Conversely, PO treatment increased anti-inflammatory genes (Mrc1, Tgfb1, Il10, Mgl2) and oxidative metabolism, characteristic of M2 macrophages. Therefore, saturated and unsaturated fatty acids bring about opposite macrophage polarization states. Coincubation of BMDM with both fatty acids counteracted the PA-induced Nos2 expression in a PO dose-dependent fashion. PO also prevented PA-induced IκBα degradation, RelA nuclear translocation, NO production, and cytokine secretion. Mechanistically, PO exerted its anti-inflammatory function through AMP-activated protein kinase as AMP kinase knockout or inhibition by Compound C offset the PO-dependent prevention of PA-induced inflammation. These results demonstrate a nutritional memory of BMDM ex vivo, highlight the plasticity of BMDM polarization in response to saturated and unsaturated fatty acids, and identify the potential to reverse diet- and saturated fat-induced M1-like polarization by administering palmitoleate. These findings could have applicability to reverse obesity-linked inflammation in metabolically relevant tissues.


Subject(s)
AMP-Activated Protein Kinases/immunology , Cell Polarity , Fatty Acids, Monounsaturated/immunology , Macrophages/immunology , Obesity/enzymology , Obesity/immunology , AMP-Activated Protein Kinases/genetics , Animals , Diet, High-Fat/adverse effects , Dietary Fats/adverse effects , Dietary Fats/metabolism , Fatty Acids, Monounsaturated/chemistry , Fatty Acids, Monounsaturated/metabolism , Humans , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/metabolism
16.
Clin Exp Immunol ; 177(2): 373-80, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24547942

ABSTRACT

Although primary biliary cirrhosis (PBC) is considered a model autoimmune disease, it has not responded therapeutically to traditional immunosuppressive agents. In addition, PBC may recur following liver transplantation, despite the absence of major histocompatibility complex (MHC) matching, in sharp contrast to the well-known paradigm of MHC restriction. We have suggested previously that invariant natural killer T (iNK T) cells are critical to the initiation of PBC. In this study we have taken advantage of our ability to induce autoimmune cholangitis with 2-octynoic acid, a common component of cosmetics, conjugated to bovine serum albumin (2-OA-BSA), and studied the natural history of pathology in mice genetically deleted for CD4 or CD8 following immunization with 2-OA-BSA in the presence or absence of α-galactosylceramide (α-GalCer). In particular, we address whether autoimmune cholangitis can be induced in the absence of traditional CD4 and CD8 responses. We report herein that CD4 and CD8 knock-out mice immunized with 2-OA-BSA/PBS or 2-OA-BSA/α-GalCer develop anti-mitochondrial antibodies (AMAs), portal infiltrates and fibrosis. Indeed, our data suggest that the innate immunity is critical for immunopathology and that the pathology is exacerbated in the presence of α-GalCer. In conclusion, these data provide not only an explanation for the recurrence of PBC following liver transplantation in the absence of MHC compatibility, but also suggest that effective therapies for PBC must include blocking of both innate and adaptive pathways.


Subject(s)
Autoimmune Diseases/immunology , Cholangitis/immunology , Immunity, Innate , Animals , Autoantibodies/blood , Autoantibodies/immunology , Autoimmune Diseases/chemically induced , Autoimmune Diseases/genetics , CD4 Antigens/genetics , CD4 Antigens/immunology , CD8 Antigens/genetics , CD8 Antigens/immunology , Cholangitis/chemically induced , Cholangitis/genetics , Dihydrolipoyllysine-Residue Acetyltransferase/immunology , Disease Models, Animal , Fatty Acids, Monounsaturated/adverse effects , Fatty Acids, Monounsaturated/immunology , Female , Galactosylceramides/administration & dosage , Galactosylceramides/adverse effects , Liver/immunology , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Biliary/immunology , Mice , Mice, Knockout , Mitochondrial Proteins/immunology , Serum Albumin, Bovine/adverse effects , Serum Albumin, Bovine/immunology , Xenobiotics/adverse effects
18.
PLoS One ; 8(9): e74225, 2013.
Article in English | MEDLINE | ID: mdl-24040208

ABSTRACT

Primary biliary cirrhosis (PBC) is considered a model autoimmune disease, with the most highly directed and specific autoantibody in both murine and human autoimmunity, the anti-mitochondrial autoantibody (AMA). However, therapeutic advances in this disease have lagged behind. Herein we have taken advantage of our unique model of murine PBC in which mice immunized with 2-octynoic acid coupled to BSA (2OA-BSA), a compound identified by quantitative structure activity relationships (QSAR) of human AMA binding, develop an intense inflammatory cholangitis with striking similarities to humans with PBC. In particular, we have constructed several unique gene-deleted mice, including mice deleted of IL-12p40, IL-12p35, IFN-γ, IL-23p19, IL-17A, IL-17F and IL-22, immunized these animals with 2OA-BSA and followed the natural history of immunopathology to identify key pathways that might provide clues for successful therapy. Our data indicate that whereas both IL-12/Th1 and IL-23/Th17 are involved in cholangitis, it is the IL-12/Th1 signaling pathway that elicits pathology. In fact, deletion of IFN-γ prevents disease and suppresses autoantibodies. Importantly, deletion of the Th17 cytokines IL-17A and IL-22, but not IL-17F, reduces biliary damage; IL-17A-knockout mice have reduced levels of anti-mitochondrial antibody. We further demonstrate that the production of IFN-γ is significantly decreased in the liver of IL-23p19(-/-), IL-17A(-/-) and IL-22(-/-) mice compared with controls. However, the ability of T cells to produce IFN-γ was not affected in Th17 cytokine-deficient mice. Our data indicate that a deficient Th17 pathway suppresses the accumulation of IFN-γ producing cells in liver during the early phase of cholangitis. In conclusion, whereas IFN-γ has a pivotal role in the early events involved in the pathogenesis of autoimmune cholangitis induced by 2OA-BSA, the IL-23/Th17 pathway potentiates the effects of IL-12/IFN-γ-mediated immunopathology.


Subject(s)
Autoantibodies/biosynthesis , Cholangitis/genetics , Interleukin-12 Subunit p35/immunology , Liver Cirrhosis, Biliary/genetics , Liver/metabolism , Signal Transduction/immunology , Animals , Autoimmunity , Cholangitis/chemically induced , Cholangitis/immunology , Cholangitis/pathology , Disease Models, Animal , Fatty Acids, Monounsaturated/chemistry , Fatty Acids, Monounsaturated/immunology , Gene Expression Regulation , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/immunology , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-12 Subunit p35/deficiency , Interleukin-12 Subunit p35/genetics , Interleukin-17/deficiency , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-23 Subunit p19/deficiency , Interleukin-23 Subunit p19/genetics , Interleukin-23 Subunit p19/immunology , Interleukins/deficiency , Interleukins/genetics , Interleukins/immunology , Liver/immunology , Liver/pathology , Liver Cirrhosis, Biliary/chemically induced , Liver Cirrhosis, Biliary/immunology , Liver Cirrhosis, Biliary/pathology , Mice , Mice, Knockout , Mitochondria/immunology , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/immunology , Th1-Th2 Balance , Interleukin-22
19.
Mol Ther ; 20(2): 356-66, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22031237

ABSTRACT

Short interfering RNA (siRNA) is a potent activator of the mammalian innate immune system. When considering possible clinical applications of siRNA for humans, the adverse immunostimulatory effects must also be taken into account. Here, we show that atelocollagen-mediated systemic delivery of siRNA without chemical modifications did not cause any immunostimulation in both animals and human peripheral blood mononuclear cells (PBMCs), even if the siRNA harbored an interferon (IFN)-inducible sequence. In contrast, systemic delivery of immunostimulatory RNA (isRNA)-mediated by a cationic lipid (such as Invivofectamine) induced potent type-I IFNs and inflammatory cytokines. Regarding the mechanism by which the isRNA/atelocollagen complex avoided adverse effects on immunostimulation, we revealed that this complex was not incorporated into PBMCs. On the other hand, Invivofectamine delivered isRNA into PBMCs. The use of either atelocollagen or Invivofectamine as a vehicle elicited significant and undistinguishable therapeutic effects in a contact hypersensitivity (CHS) inflammatory model mouse, when we intravenously injected the siRNA targeting monocyte chemoattractant protein-1 as the complex. For the goal of realizing siRNA-based medicines for humans, atelocollagen is an excellent and promising delivery vehicle, and it has the useful advantage of evading detection by the "radar" of innate immunity.


Subject(s)
Collagen/administration & dosage , RNA, Small Interfering/administration & dosage , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/toxicity , Animals , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Dermatitis, Contact/immunology , Dermatitis, Contact/therapy , Disease Models, Animal , Fatty Acids, Monounsaturated/chemistry , Fatty Acids, Monounsaturated/immunology , Female , Gene Transfer Techniques , Humans , Inflammation Mediators/metabolism , Interferon Type I/biosynthesis , Kidney Function Tests , Leukocytes, Mononuclear/immunology , Liver/enzymology , Macrophages/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred ICR , Quaternary Ammonium Compounds/chemistry , Quaternary Ammonium Compounds/immunology , RNA, Small Interfering/chemistry , RNA, Small Interfering/toxicity , Receptors, Cell Surface/metabolism
20.
Clin Exp Immunol ; 166(1): 110-20, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21910728

ABSTRACT

A void in understanding primary biliary cirrhosis (PBC) is the absence of appropriate animal models. Our laboratory has studied a murine model of autoimmune cholangitis induced following immunization with 2-octynoic acid (2OA), an antigen identified following extensive quantitative structural activity relationship (QSAR) analysis, using human autoantibodies and three-dimensional analysis of the mitochondrial autoantigen, the E2 subunit of the pyruvate dehydrogenase complex (PDC-E2). Mice immunized with 2OA coupled to bovine serum albumin (BSA) develop anti-mitochondrial antibodies (AMAs) of the identical specificity as humans with PBC, and in addition develop inflammatory portal cell infiltrates in liver. However, the natural history of disease is less severe than in humans and does not include fibrosis. Data from human and autoimmune murine models suggest that environmental and/or infectious agents can exacerbate autoimmune reactions, and a model of PBC has been described in which polyinosinic-polycytidylic acid (poly I:C), a viral RNA mimetic and Toll-like receptor 3 (TLR-3) agonist induces low-titre AMAs and in mild portal infiltrates. We took advantage of our established model to determine whether immunization with 2OA-BSA coupled with poly I:C alters the disease process. Indeed, the addition of poly I:C produces a profound exacerbation of autoimmune cholangitis, including a significant increase in CD8(+) infiltrating T cells, as well as a marked increase of proinflammatory cytokines. In addition, mice have evidence of fibrosis. These findings lend support to the concept that besides breakdown of self-tolerance, there is a requirement of a second 'hit' during the breakdown process that leads to disease which more faithfully mimics human PBC.


Subject(s)
Autoimmune Diseases/immunology , Cholangitis/immunology , Dihydrolipoyllysine-Residue Acetyltransferase/adverse effects , Disease Models, Animal , Fatty Acids, Monounsaturated/adverse effects , Liver Cirrhosis, Biliary/immunology , Liver/immunology , Mitochondrial Proteins/adverse effects , Poly I-C/adverse effects , Toll-Like Receptor 3/immunology , Animals , Autoantibodies/blood , Autoantibodies/immunology , Autoantigens/adverse effects , Autoantigens/chemistry , Autoantigens/immunology , Autoimmune Diseases/chemically induced , Autoimmune Diseases/pathology , Cattle , Cholangitis/chemically induced , Cholangitis/pathology , Cytokines/biosynthesis , Cytokines/immunology , Dihydrolipoyllysine-Residue Acetyltransferase/chemistry , Dihydrolipoyllysine-Residue Acetyltransferase/immunology , Fatty Acids, Monounsaturated/chemistry , Fatty Acids, Monounsaturated/immunology , Female , Humans , Immunization , Liver/drug effects , Liver/pathology , Liver Cirrhosis, Biliary/chemically induced , Liver Cirrhosis, Biliary/pathology , Male , Mice , Mice, Inbred C57BL , Mitochondria/immunology , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/immunology , Poly I-C/chemistry , Poly I-C/immunology , Serum Albumin/chemistry , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...