Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Ocul Immunol Inflamm ; : 1-15, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38127798

ABSTRACT

PURPOSE: To explore the role of the proinflammatory cytokine, macrophage migration inhibitory factor (MIF), in a murine model of dry eye disease (DED). METHODS: The role of MIF on DED was determined using genetically MIF deficient mice and pharmacological inhibition of MIF. DED was induced with 0.5 mg of scopolamine via subcutaneous injection in wild type (WT) and mice lacking MIF (Mif-/-), three times a day for 21 days. DED signs, tear volume, ferning pattern and cytology impression were evaluated. Also, eye tissues were collected to determine transcripts of key inflammatory mediators and histopathological damage. In a second set of experiments, we neutralized MIF with ISO-1, an isozaxiline-derivative MIF tautomerase activity-inhibiting small molecule in WT mice, following an acute DED model for 10 days. ISO-1 was given starting on day 3 after DED induction and signs were evaluated, including a recovery phase in both experimental approaches. RESULTS: When compared to WT, Mif-/- mice showed attenuated signs of DED like preserved mucin pattern and increased tear volume. Also, Mif-/- mice maintained conjunctival epithelial cells and less corneal damage, associated with lower levels of TNFα and IL-1ß. At recovery phase, Mif-/- mice presented improved signs. Interestingly, in cornea and conjunctiva the absence of MIF selectively downregulated the transcription of inflammatory enzymes like inos and nox4 whereas displayed enhanced transcripts of il-4, il-13, tgfß and cox2. Finally, pharmacological inhibition of MIF using ISO-1, replicated the above findings in the mouse model. CONCLUSION: MIF is a central positive mediator of the inflammatory process in experimental DED, thus, targeting MIF could be used as a novel therapy in ocular surface inflammatory pathologies.

2.
Microbes Infect ; 17(8): 586-95, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25899946

ABSTRACT

Infection of C57BL/6J mice with the parasite Toxoplasma gondii triggers a powerful Th1 immune response that is detrimental to the host. During acute infection, a reduction in CD4(+)Foxp3(+) regulatory T cells (Treg) has been reported. We studied the role of Treg during T. gondii infection by adoptive transfer of cells purified from transgenic Foxp3(EGFP) mice to infected wild type animals. We found a less severe weight loss, a significant delayed mortality in infected Treg-transferred mice, and reduced pathology of the small intestine that were associated with lower IFN-γ and TNF-α levels. Nevertheless, higher cyst number and parasite load in brain were observed in these mice. Treg-transferred infected mice showed reduced levels of both IFN-γ and TNF-α in sera. A reduced number of CD4(+) T cells producing IFN-γ was detected in these mice, while IL-2 producing CD4(+) T cells were restored to levels nearly similar to uninfected mice. CD25 and CD69 expression of CD4(+) T cells were also down modulated. Our data show that the low Treg cell number are insufficient to modulate the activation of CD4(+) T cells and the production of high levels of IFN-γ. Thus, a delicate balance between an optimal immune response and its modulation by Treg cells must exist.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/metabolism , Interleukin-2/metabolism , Th1 Cells/immunology , Toxoplasmosis/immunology , Acute Disease , Animals , Down-Regulation/immunology , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Toxoplasmosis/metabolism , Toxoplasmosis/pathology
3.
Int J Biol Sci ; 10(8): 909-20, 2014.
Article in English | MEDLINE | ID: mdl-25170304

ABSTRACT

The C-type lectin receptor mMGL is expressed exclusively by myeloid antigen presenting cells (APC) such as dendritic cells (DC) and macrophages (Mφ), and it mediates binding to glycoproteins carrying terminal galactose and α- or ß-N-acetylgalactosamine (Gal/GalNAc) residues. Trypanosoma cruzi (T. cruzi) expresses large amounts of mucin (TcMUC)-like glycoproteins. Here, we show by lectin-blot that galactose moieties are also expressed on the surface of T. cruzi. Male mMGL knockout (-/-) and wild-type (WT) C57BL/6 mice were infected intraperitoneally with 10(4) T. cruzi trypomastigotes (Queretaro strain). Following T. cruzi infection, mMGL-/- mice developed higher parasitemia and higher mortality rates compared with WT mice. Although hearts from T. cruzi-infected WT mice presented few amastigote nests, mMGL-/- mice displayed higher numbers of amastigote nests. Compared with WT, Mφ from mMGL-/- mice had low production of nitric oxide (NO), interleukin (IL)-12 and tumor necrosis factor (TNF)-α in response to soluble T. cruzi antigens (TcAg). Interestingly, upon in vitro T. cruzi infection, mMGL-/- Mφ expressed lower levels of MHC-II and TLR-4 and harbored higher numbers of parasites, even when mMGL-/- Mφ were previously primed with IFN-γ or LPS/IFN-γ. These data suggest that mMGL plays an important role during T. cruzi infection, is required for optimal Mφ activation, and may synergize with TLR-4-induced pathways to produce TNF-α, IL-1ß and NO during the early phase of infection.


Subject(s)
Galactose/metabolism , Lectins, C-Type/metabolism , Macrophages/metabolism , Trypanosoma cruzi/physiology , Trypanosomiasis/immunology , Animals , Immunity/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Biomed Res Int ; 2013: 316980, 2013.
Article in English | MEDLINE | ID: mdl-23509709

ABSTRACT

It was previously reported by our group that infection with Taenia crassiceps reduces incidence and severity of inflammatory and autoimmune experimental diseases like type 1 diabetes and experimental autoimmune encephalomyelitis. In this research, we set out to study whether infection with T. crassiceps would affect the development of experimental rheumatoid arthritis (RA). We found that mice infected with the parasite and induced with experimental RA showed similar clinical scores as the noninfected experimental RA group; systemic cytokines were not affected while anti-CII Abs were higher in the infected group. Histological evaluation showed damage in both infected and noninfected experimental RA-induced groups and although some surface molecules such as PDL-2 and MR which are associated with immunomodulatory mechanisms were upregulated in the infected and RA-induced group as compared to the noninfected RA group, they did not exert any changes in the outcome of experimental RA. Thus, we determined that infection with T. crassiceps does not influence the outcome of experimental RA.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Taeniasis/immunology , Animals , Arthritis, Experimental/complications , Arthritis, Experimental/parasitology , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/parasitology , Autoimmune Diseases/immunology , Cell Membrane/metabolism , Disease Models, Animal , Immunoglobulin G/immunology , Inflammation , Interferon-gamma/metabolism , Macrophages, Peritoneal/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Taenia , Taeniasis/complications
5.
Int J Biol Sci ; 7(9): 1298-310, 2011.
Article in English | MEDLINE | ID: mdl-22110382

ABSTRACT

Macrophage migration inhibitory factor (MIF) has been found to be involved in host resistance to several parasitic infections. To determine the mechanisms of the MIF-dependent responses to Trypanosoma cruzi, we investigated host resistance in MIF⁻/⁻ mice (on the BALB/c background) during an intraperitoneal infection. We focused on the potential involvement of MIF in dendritic cell (DC) maturation and cytokine production. Following a challenge with 5 x 10(3)T. cruzi parasites, wild type (WT) mice developed a strong IL-12 response and adequate maturation of the draining mesenteric lymph node DCs and were resistant to infection. In contrast, similarly infected MIF⁻/⁻ mice mounted a weak IL-12 response, displayed immature DCs in the early phases of infection and rapidly succumbed to T. cruzi infection. The lack of maturation and IL-12 production by the DCs in response to total T. cruzi antigen (TcAg) was confirmed by in vitro studies. These effects were reversed following treatment with recombinant MIF. Interestingly, TcAg-stimulated bone marrow-derived DCs from both WT and MIF⁻/⁻ mice had increased ERK1/2 MAPK phosphorylation. In contrast, p38 phosphorylation was only upregulated in WT DCs. Reconstitution of MIF to MIF⁻/⁻ DCs upregulated p38 phosphorylation. The MIF-p38 pathway affected MHC-II and CD86 expression as well as IL-12 production. These findings demonstrate that the MIF-induced early DC maturation and IL-12 production mediates resistance to T. cruzi infection, probably by activating the p38 pathway.


Subject(s)
Antigens, Protozoan/metabolism , Interleukin-12/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Animals , Antigens, Protozoan/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Interferon-gamma/metabolism , Interleukin-1beta/metabolism , Macrophage Migration-Inhibitory Factors/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Phosphorylation , Tumor Necrosis Factor-alpha/metabolism
6.
FASEB J ; 24(7): 2583-90, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20203087

ABSTRACT

Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine involved in the pathogenesis of a variety of autoimmune inflammatory diseases. Here, we investigated the role of MIF in the pathogenesis of non-insulin-dependent diabetes mellitus (NIDDM) using MIF(-/-) mice and a mouse model of streptozotocin (STZ)-induced NIDDM. Following single injection of STZ, MIF(+/+) BALB/c mice showed a significant increase in blood glucose levels, developed polyuria, and succumbed to disease. In contrast, no such increase in blood glucose was observed in MIF(-/-) BALB/c mice treated with STZ. These mice produced significantly less inflammatory cytokines and resistin as compared with MIF(+/+) mice and failed to develop clinical disease. Finally, oral administration of a small-molecule MIF antagonist, CPSI-1306, to outbred ICR mice following induction of NIDDM significantly lowered blood glucose levels in the majority of animals, which was also associated with a significant reduction in the levels of the proinflammatory cytokines IL-6 and TNF-alpha in the sera. Taken together, these results demonstrate that MIF is involved in the pathogenesis of NIDDM and is a therapeutic target to treat this disease.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Macrophage Migration-Inhibitory Factors/deficiency , Animals , Blood Glucose , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/etiology , Female , Interleukin-6/blood , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/physiology , Mice , Mice, Knockout , Treatment Outcome , Tumor Necrosis Factor-alpha/blood
7.
Exp Parasitol ; 126(3): 348-58, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20331989

ABSTRACT

Macrophage migration inhibitory factor (MIF) has been found to be involved in host resistance to several parasitic infections. To determine the mechanisms of MIF-dependent responses to Toxoplasma gondii, we investigated host resistance in MIF-/- mice (BALB/c background) during natural oral infection. We focused on the potential involvement of MIF in Dendritic Cell (DC) maturation and IL-12 production. Following oral T. gondii infection, wild type mice developed a strong IL-12 response with an adequate maturation of their draining mesenteric lymph node DC (MLNDC) population and were resistant to challenge with either 40 or 100 cysts (ME49 strain). In contrast, similarly infected MIF-/- mice mounted a weak IL-12 response, displayed immature MLNDCs in the early phases of infection and rapidly succumbed to both type of challenges. Lack of maturation and IL-12 production of DCs in response to T. gondii antigens was confirmed by in vitro studies, and these effects were reversed following treatment with recombinant MIF. These findings demonstrate that MIF-induced early DC maturation and IL-12 production mediate resistance to T. gondii infection.


Subject(s)
Dendritic Cells/pathology , Interleukin-12/biosynthesis , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Toxoplasmosis, Animal/immunology , Animals , Antigens, Protozoan/immunology , Brain/parasitology , Dendritic Cells/immunology , Disease Progression , Disease Susceptibility/immunology , Histocompatibility Antigens Class II/metabolism , Intramolecular Oxidoreductases/physiology , Liver/parasitology , Liver/pathology , Macrophage Migration-Inhibitory Factors/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Specific Pathogen-Free Organisms , Toxoplasma/genetics , Toxoplasma/growth & development , Toxoplasma/immunology , Up-Regulation
8.
J Biomed Biotechnol ; 2010: 505694, 2010.
Article in English | MEDLINE | ID: mdl-20111744

ABSTRACT

The aryl hydrocarbon receptor (AhR) is part of a signaling system that is mainly triggered by xenobiotic agents. Increasing evidence suggests that AhR may regulate immunity to infections. To determine the role of AhR in the outcome of toxoplasmosis, we used AhR-/- and wild-type (WT) mice. Following an intraperitoneal infection with Toxoplasma gondii (T. gondii), AhR-/- mice succumbed significantly faster than WT mice and displayed greater liver damage as well as higher serum levels of tumor necrosis factor (TNF)-alpha, nitric oxide (NO), and IgE but lower IL-10 secretion. Interestingly, lower numbers of cysts were found in their brains. Increased mortality was associated with reduced expression of GATA-3, IL-10, and 5-LOX mRNA in spleen cells but higher expression of IFN-gamma mRNA. Additionally, peritoneal exudate cells from AhR-/- mice produced higher levels of IL-12 and IFN-gamma but lower TLR2 expression than WT mice. These findings suggest a role for AhR in limiting the inflammatory response during toxoplasmosis.


Subject(s)
Receptors, Aryl Hydrocarbon/immunology , Toxoplasma/immunology , Toxoplasmosis, Animal/immunology , Animals , Antigens, Protozoan/immunology , Ascitic Fluid/metabolism , Brain/immunology , Female , Flow Cytometry , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Immunity, Innate/immunology , Inbreeding , Interferon-gamma/biosynthesis , Interferon-gamma/blood , Interferon-gamma/genetics , Interleukins/biosynthesis , Interleukins/blood , Lipoxins/biosynthesis , Lipoxins/genetics , Liver/immunology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Spleen/immunology , Toxoplasmosis, Animal/metabolism , Toxoplasmosis, Animal/pathology , Tumor Necrosis Factor-alpha/blood
9.
FASEB J ; 22(10): 3661-71, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18606868

ABSTRACT

Macrophage migration inhibitory factor (MIF) exerts either a protective or a deleterious role in the immune response to different pathogens. We analyzed herein the role of MIF in the host control of toxoplasmosis using MIF(-/-) mice backcrossed to either the BALB/c or the C57BL/6 genetic backgrounds. Both, wild-type (WT) BALB/c and MIF(-/-) BALB/c mice were susceptible to infection with highly virulent RH as well as moderately virulent ME49 strains of T. gondii. MIF(-/-) mice, however, showed greater liver damage and more brain cysts, produced less proinflammatory cytokines, and succumbed significantly faster than WT mice. Bone marrow-derived dendritic cells (BMDCs) from MIF(-/-) mice produced less interleukin-1beta, interleukin-12, and tumor necrosis factor-alpha than WT BMDCs after stimulation with soluble Toxoplasma antigen (STAg). Similar observations were made in CD11c(+) low-density cells isolated from the spleens of MIF(-/-) mice challenged with STAg. MIF(-/-) C57BL/6 mice succumbed to ME49 infection faster than their WT counterparts. C57BL/6 mice that succumbed to infection with the ME49 strain produced less MIF than resistant BALB/c mice similarly infected. Interestingly, an analysis of brains from patients with cerebral toxoplasmosis showed low levels of MIF expression. Together, these findings demonstrate that MIF plays a critical role in mediating host resistance against T. gondii.


Subject(s)
Genetic Predisposition to Disease , Host-Parasite Interactions , Immunity, Innate , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/physiology , Toxoplasmosis/genetics , Animals , Brain/metabolism , Brain/parasitology , Cytokines/biosynthesis , Dendritic Cells/immunology , Encephalitis/immunology , Encephalitis/parasitology , Encephalitis/pathology , Hepatitis/immunology , Hepatitis/parasitology , Hepatitis/pathology , Host-Parasite Interactions/genetics , Host-Parasite Interactions/immunology , Humans , Immunity, Innate/genetics , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Mice , Mice, Mutant Strains , Nitric Acid/metabolism , Toll-Like Receptor 2/biosynthesis , Toxoplasma/pathogenicity , Toxoplasmosis/immunology , Toxoplasmosis/pathology , Toxoplasmosis, Cerebral/immunology , Toxoplasmosis, Cerebral/parasitology , Toxoplasmosis, Cerebral/pathology , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...