Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Infect Immun ; 86(5)2018 05.
Article in English | MEDLINE | ID: mdl-29531131

ABSTRACT

Interleukin-10 (IL-10)-producing regulatory B (Breg) cells were found to be induced in a variety of infectious diseases. However, its importance in the regulation of immune response to malaria is still unclear. Here, we investigated the dynamics, phenotype, and function of Breg cells using Plasmodium chabaudi chabaudi AS-infected C57BL/6 and BALB/c mice. BALB/c mice were more susceptible to infection and had a stronger IL-10 response in spleen than C57BL/6 mice. Analysis of the surface markers of IL-10-producing cells with flow cytometry showed that CD19+ B cells were one of the primary IL-10-producing populations in P. c. chabaudi AS-infected C57BL/6 and BALB/c mice, especially in the latter one. The Breg cells had a heterogeneous phenotype which shifted during infection. The well-established Breg subset, CD19+ CD5+ CD1dhi cells, accounted for less than 20% of IL-10-producing B cells in both strains during the course of infection. Most Breg cells were IgG+ and CD138- from day 0 to day 8 postinfection. Adoptive transfer of Breg cells to C57BL/6 mice infected with P. c. chabaudi AS led to a transient increase of parasitemia without an impact on survival rate. Our finding reveals that B cells play an active and important regulatory role in addition to mediating humoral immunity in immune response against malaria, which should be paid more attention in developing therapeutic or vaccine strategies against malaria involving stimulation of B cells.


Subject(s)
B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/metabolism , Interleukin-10/immunology , Interleukin-10/metabolism , Malaria/immunology , Plasmodium chabaudi/immunology , Spleen/immunology , Spleen/metabolism , Adoptive Transfer , Animals , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
2.
J Immunol ; 193(3): 1314-23, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24965778

ABSTRACT

In animal models of experimental cerebral malaria (ECM), neuropathology is associated with an overwhelming inflammatory response and sequestration of leukocytes and parasite-infected RBCs in the brain. In this study, we explored the effect of vitamin D (VD; cholecalciferol) treatment on host immunity and outcome of ECM in C57BL/6 mice during Plasmodium berghei ANKA (PbA) infection. We observed that oral administration of VD both before and after PbA infection completely protected mice from ECM. VD administration significantly dampened the inducible systemic inflammatory responses with reduced circulating cytokines IFN-γ and TNF and decreased expression of these cytokines by the spleen cells. Meanwhile, VD also resulted in decreased expression of the chemokines CXCL9 and CXCL10 and cytoadhesion molecules (ICAM-1, VCAM-1, and CD36) in the brain, leading to reduced accumulation of pathogenic T cells in the brain and ultimately substantial improvement of the blood-brain barriers of PbA-infected mice. In addition, VD inhibited the differentiation, activation, and maturation of splenic dendritic cells. Meanwhile, regulatory T cells and IL-10 expression levels were upregulated upon VD treatment. These data collectively demonstrated the suppressive function of VD on host inflammatory responses, which provides significant survival benefits in the murine ECM model.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Immunosuppressive Agents/administration & dosage , Malaria, Cerebral/immunology , Malaria, Cerebral/prevention & control , Plasmodium berghei/immunology , Vitamin D/administration & dosage , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Dendritic Cells/immunology , Dendritic Cells/pathology , Down-Regulation/immunology , Female , Growth Inhibitors/administration & dosage , Growth Inhibitors/therapeutic use , Host-Parasite Interactions/immunology , Immunosuppressive Agents/therapeutic use , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Malaria, Cerebral/pathology , Mice , Mice, Inbred C57BL , Primary Cell Culture , Random Allocation , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation/immunology , Vitamin D/therapeutic use
3.
Exp Parasitol ; 170: 73-81, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27646627

ABSTRACT

The mechanisms regulating the induction of protective immunity against blood-stage malaria remain unclear. Resistant DBA/2 mouse develops a higher Th1 response compared with a susceptible BALB/c strain during Plasmodium yoelii (Py) infection. It is known that the T helper cell response is initiated and polarized by dendritic cells (DCs) of the innate immune system, during which TLR4 and TLR9 are important receptors for the innate recognition of the malaria parasite and its products. We hypothesized that TLR4/9 may play critical roles in the induction of protective immunity against Py infection. We used TLR4/9 antagonists and agonists to study their effects on mouse resistance to Py infection. We found that the administration of an antagonist prior to infection aggravated disease outcomes, impaired DC functions and suppressed the pro-inflammatory response to Py infection in resistant DBA/2 mice. Treatment with the TLR4 agonist lipopolysaccharide (LPS) but not TLR9 agonist significantly improved the survival rate of susceptible Py-infected BALB/c mice. LPS administration promoted the activation and expansion of DCs and drove a Th1-biased response. Our data demonstrate the important roles of TLR4/9 signals in inducing resistance to malaria parasites and provide evidence for the rational use of TLR agonists to potentiate protective immunity against Plasmodium infection.


Subject(s)
Malaria/immunology , Plasmodium yoelii/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 9/immunology , Animals , CD4-Positive T-Lymphocytes/drug effects , Cells, Cultured , Cytokines/drug effects , Dendritic Cells/drug effects , Dendritic Cells/physiology , Female , Flow Cytometry , Interferon-gamma/analysis , Interleukin-10/analysis , Lipopolysaccharides/pharmacology , Malaria/parasitology , Malaria/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Oligodeoxyribonucleotides/pharmacology , Parasitemia/immunology , Parasitemia/parasitology , Signal Transduction/immunology , Spleen/cytology , Spleen/immunology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/antagonists & inhibitors
4.
Exp Parasitol ; 156: 1-11, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26005191

ABSTRACT

Phenylhydrazine (PHZ) treatment is generally used to enhance parasitemia in infected mice models. Transient reticulocytosis is commonly observed in iron-deficient anemic hosts after treatment with iron supplementation, and is also associated with short-term hemolysis caused by PHZ treatment. In this study, we investigated the relationship between reticulocytosis and cerebral malaria (CM) in a murine model induced by PHZ administration before Plasmodium berghei ANKA (PbA) infection. Mortality and parasitemia were checked daily. Pro-inflammatory cytokines and IL-10 were quantified by ELISA. The expression of CXCL9, CXCL10, CCL5, and CXCR3 mRNAs was determined by real-time PCR. Brain sequestration of CD4(+) and CD8(+) T cells and populations of splenic Th1 CD4(+) T cells, dendritic cells (DCs), CD11b(+) Gr1(+) cells, and regulatory T cells (Tregs) were assessed by FACS. PHZ administration dramatically increased parasitemia from day 3 to day 5 post infection (p.i.) compared with the untreated control infected mice group; also, CM developed at day 5 p.i., compared with day 7 p.i. in untreated control infected mice, as well as significantly decreased blood-brain barrier function (P < 0.001). PHZ administration during PbA infection significantly increased the expression of CXCL9 (P <0.05) and VCAM-1 (P <0.001) in the brain, increased the expression of CXCL10, CCL5 and CXCR3, and significantly increased the recruitment of CD4(+) and CD8(+) T cells (P <0.001 and P <0.01, respectively) as well as CD11b(+) Gr1(+) cells to the brain. In addition, PHZ administration significantly increased the numbers of IL-12-secreting DCs at days 3 and 5 p.i. compared to those of untreated control infected mice (P <0.001 and P <0.01, respectively). Consequently, the activation of CD4(+) T cells, especially the expansion of the Th1 subset (P <0.05), was significantly and dramatically enhanced and was accompanied by marked increases in the production of protein and/or mRNA of the Th1-type pro-inflammatory mediators, IFN-γ and TNF-α (P <0.01 for both for protein; P <0.05 for TNF-α mRNA). Our results suggest that, compared to healthy individuals, people suffering from reticulocytosis may be more susceptible to severe malaria infection in malaria endemic areas. This has implications for the most appropriate selection of treatment, which may also cause reticulocytosis in patients living in such areas.


Subject(s)
Malaria, Cerebral/chemically induced , Oxidants/adverse effects , Parasitemia/chemically induced , Phenylhydrazines/adverse effects , Plasmodium berghei/drug effects , Reticulocytosis/drug effects , Animals , Blood-Brain Barrier/metabolism , Erythrocyte Count , Erythrocyte Indices , Female , Hemoglobins/analysis , Injections, Intraperitoneal , Mice , Mice, Inbred C57BL , Plasmodium berghei/pathogenicity , Random Allocation , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism , Reticulocytes/cytology , Reticulocytes/drug effects , Reticulocytosis/immunology , Spleen/cytology , Spleen/immunology , T-Lymphocytes/drug effects , Up-Regulation
5.
Tohoku J Exp Med ; 236(1): 21-31, 2015 05.
Article in English | MEDLINE | ID: mdl-25925198

ABSTRACT

L-Arginine (L-Arg), the substrate for nitric oxide (NO) synthase, has been used to treat malaria to reverse endothelial dysfunction in adults. However, the safety and efficacy of L-Arg remains unknown in malaria patients under the age of five, who are at the greatest risk of developing cerebral malaria (CM), a severe malaria complication. Here, we tested effects of L-Arg treatment on the outcomes of CM using a mouse model. Experimental cerebral malaria (ECM) was induced in female C57BL/6 mice infected with Plasmodium berghei ANKA, and L-Arg was administrated either prophylactically or after parasite infection. Surprisingly, both types of L-Arg administration caused a decline in survival time and raised CM clinical scores. L-Arg treatment increased the population of CD4(+)T-bet(+)IFN-γ(+) Th1 cells and the activated macrophages (F4/80(+)CD36(+)) in the spleen. The levels of pro-inflammatory cytokines, IFN-γ and TNF-α, in splenocyte cultures were also increased by L-Arg treatment. The above changes were accompanied with a rise in the number of dendritic cells (DCs) and an increase in their maturation. However, L-Arg did not affect the population of regulatory T cells or the level of IL-10 in the spleen. Taken together, these data suggest that L-Arg may enhance the Th1 immune response, which is essential for a protective response in uncomplicated malaria but could be lethal in CM patients. Therefore, the prophylactic use of L-Arg to treat CM, based on the assumption that restoring the bioavailability of endothelial NO improves the outcome of CM, may need to be reconsidered especially for children.


Subject(s)
Arginine/adverse effects , Disease Progression , Inflammation Mediators/metabolism , Malaria, Cerebral/pathology , Animals , Cell Proliferation/drug effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Immunity/drug effects , Interleukin-10/metabolism , Lymphocyte Count , Macrophage Activation/drug effects , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Parasitemia/immunology , Phenotype , Plasmodium berghei/drug effects , Plasmodium berghei/immunology , Survival Analysis , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th1 Cells/drug effects , Th1 Cells/immunology
6.
Immunogenetics ; 66(4): 231-42, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24566892

ABSTRACT

Inbred stains of mice display differential susceptibility to infection with the common foodborne pathogen Listeria monocytogenes (Lm). Previously, Listr1 and Listr2, two genetic loci that control differential sensitivity to Lm infection between BALB/cByJ and C57BL/6ByJ mice, were identified. To analyze the role of Listr1 in innate immune responses, we employed congenic mice (C.B6By-Listr1/Rag2 (-/-) ) bearing the C57BL/6ByJ-derived Listr1 locus on a BALB/c-Rag2 (-/-) background. Consistent with the results of a previous genetic analysis, the congenic mice showed increased susceptibility to Lm infection. The bacterial burden in the liver between the congenic and control lines was significantly different (P < 0.05) from 24 h postinfection with Lm. Analysis of genes within the Listr1 locus identified a frameshift mutation in the Cxcl11 gene of the C57BL/6 strain that prevents production of the mature chemokine CXCL11. No differences in inflammatory cell infiltration or cells expressing CXCR3 and CXCR7 which are the receptors of CXCL11 occurred because of CXCL11 deficiency in the congenic mice spleens. However, these mice lacked a distinct population of CD14(+) positive resident mononuclear cells that express intermediate levels of CXCR3 and CXCR7 in the liver. There were fewer microabscesses in the liver of CXCL11-deficient mice during the early stage of infection, which is consistent with their decreased ability to resist Lm. Our results, when taken together, show that the Listr1 locus plays an important role in early control of Lm infection in the mouse liver and that Cxcl11 is a candidate gene for disease severity within this locus.


Subject(s)
Chemokine CXCL11/genetics , Immunity, Innate/genetics , Listeria monocytogenes/immunology , Listeria monocytogenes/pathogenicity , Listeriosis/genetics , Listeriosis/immunology , Animals , Chemokine CXCL11/biosynthesis , Chemokine CXCL11/deficiency , Frameshift Mutation , Genetic Predisposition to Disease , Lipopolysaccharide Receptors/metabolism , Listeriosis/microbiology , Liver/immunology , Liver/microbiology , Liver/pathology , Mice , Mice, Congenic , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Polymorphism, Genetic , Receptors, CXCR/metabolism , Receptors, CXCR3/metabolism , Spleen/immunology , Spleen/pathology
7.
Malar J ; 13: 116, 2014 Mar 26.
Article in English | MEDLINE | ID: mdl-24670210

ABSTRACT

BACKGROUND: Malaria and schistosomiasis are endemic and co-exist in the same geographic areas, even co-infecting the same host. Previous studies have reported that concomitant infection with Schistosoma japonicum could offer protection against experimental cerebral malaria (ECM) in mice. This study was performed to evaluate whether alterations in parasite density could alter this protective effect. METHODS: Mice were inoculated with 100 or 200 S. japonicum cercariae followed by infection with high or low density of Plasmodium berghei ANKA strain eight weeks after the first infection. Then, parasitaemia, survival rate and blood-brain-barrier (BBB) damage were assessed. Interferon-gamma (IFN-γ), interleukin (IL)-4, IL-5, IL-13, IL-10, and TGF-ß levels were determined in splenocyte supernatants using enzyme-linked immunosorbent assay (ELISA). Cell surface/intracellular staining and flow cytometry were used to analyse the level of CD4(+)/CD8(+) T cells, CD4(+)CD25(+)Foxp3(+) Tregs, IL-10-secreting Tregs, and IL-10(+)Foxp3-CD4(+) T cells in the spleen, and CD4(+)/CD8(+) T cells infiltrating the brain. RESULTS: Co-infection with low density P. berghei and increased S. japonicum cercariae significantly increased the levels of IL-4, IL-5, IL-13, TGF-ß and Tregs, but significantly decreased the levels of IFN-γ and the percentage of CD4(+) T cells and CD8(+) T cells in the spleen and CD8(+) T cell infiltration in the brain. Increased worm loads also significantly decreased mortality and BBB impairment during ECM. When challenged with higher numbers of P. berghei and increased cercariae, the observed cytokine changes were not statistically significant. The corresponding ECM mortality and BBB impairment also remained unchanged. CONCLUSIONS: This study demonstrates that protection for ECM depends on the numbers of the parasites, S. japonicum and P. berghei, during co-infection. Alterations in the regulatory response appear to play a key role in this adaptation.


Subject(s)
Coinfection/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Schistosoma japonicum/immunology , Schistosomiasis japonica/immunology , Animals , Coinfection/parasitology , Coinfection/pathology , Cytokines/immunology , Disease Susceptibility/immunology , Disease Susceptibility/parasitology , Disease Susceptibility/pathology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immune Tolerance , Malaria, Cerebral/parasitology , Malaria, Cerebral/pathology , Mice , Mice, Inbred C57BL , Parasitemia/immunology , Parasitemia/parasitology , Parasitemia/pathology , Schistosomiasis japonica/parasitology , Schistosomiasis japonica/pathology , Spleen/immunology
8.
Tohoku J Exp Med ; 234(4): 271-80, 2014 12.
Article in English | MEDLINE | ID: mdl-25477006

ABSTRACT

Chloroquine (CQ), a well-known anti-malarial drug, has long been used for the treatment of autoimmune diseases because of its profound immunomodulatory effects. However, whether this drug modifies anti-malaria immune response is still not clear. Here we studied the immunomodulatory role of CQ in a mouse model of malaria. DBA/2 mice were infected with Plasmodium yoelii (Py) parasite (intraperitoneal injection of parasitized erythrocytes) and divided into three groups. Two groups received single dose of CQ (gavage administration) at 6 hours after Py infection (post-6h) and 3 days after Py infection (post-3d), respectively. The third group received saline as control. The course of disease was monitored and the changes of immune response were investigated. It is shown that mice from the post-6h group took longer time to clear the parasites compared with those of the post-3d group. The activation of T helper cells, macrophages, and B cells was significantly suppressed in mice with post-6h CQ treatment as compared with control mice on day 3 and day 5 after infection. In contrast, no such changes were found in mice from the post-3d group. Dendritic cells (DCs) from the post-6h CQ treated mice were less mature as compared with those from control mice as well as those from the post-3d group. Taken together, our data suggest that treatment with CQ early in infection inhibits protective immune response against Py infection possibly via mechanisms involving the modulation of DC's function. Our finding provided important information for reasonable use of CQ in malaria chemotherapy.


Subject(s)
Chloroquine/therapeutic use , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Malaria/drug therapy , Malaria/immunology , Plasmodium yoelii/immunology , Animals , Antimalarials/pharmacology , Antimalarials/therapeutic use , Chloroquine/pharmacology , Dendritic Cells/drug effects , Disease Models, Animal , Disease Progression , Female , Malaria/parasitology , Mice, Inbred DBA , Plasmodium yoelii/drug effects , Th2 Cells/drug effects , Toll-Like Receptor 9/metabolism
9.
Tohoku J Exp Med ; 229(2): 87-96, 2013 02.
Article in English | MEDLINE | ID: mdl-23303295

ABSTRACT

Listeria monocytogenes (Lm) has been used as the adjuvant or vector for tumor and viral vaccine for its capability of eliciting all aspects of cell-mediated immunity including T cell activation and interferon-gamma (IFN-γ) production. These effector components play critical roles in the protection against Plasmodium infection in both human malaria and mouse models. Therefore, immune response induced by Lm infection may benefit the defense against malaria. To test this hypothesis, we employed blood-stage Plasmodium yoelii (PyL) infected mice and challenged them with Lm. C57BL/6 and BALB/c mice that are sensitive to PyL infection were used in experiments. These two strains are resistant and sensitive, respectively, to Lm infection. The outcomes of double infection with PyL and Lm and the changes of immune response were investigated. We found that live Lm inoculation inhibited PyL multiplication in both C57BL/6 and BALB/c mice. Lm inoculation increased production of IFN-γ, infiltration of CD11b-positive macrophages and generation of nitric oxide in the spleen of C57BL/6 mice at day 5 after parasite infection. Both CD4- and CD8- positive T cells contributed to IFN-γ production induced by Lm inoculation in PyL-infected mice. The protective effect of Lm against PyL infection depended on the viability of the bacteria. Live Lm, rather than heat-killed Lm, stimulated early IFN-γ production which provided essential cytokine environment for the development of Th1 response in PyL-infected mice. Our data show for the first time that Lm inoculation has protective effect against blood-stage murine malaria, which provides a new clue for enhancing anti-Plasmodium immunity.


Subject(s)
Coinfection/immunology , Listeria monocytogenes/immunology , Malaria Vaccines/immunology , Malaria/prevention & control , Plasmodium yoelii/pathogenicity , Adjuvants, Immunologic , Animals , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Immunity, Cellular/immunology , Interferon-gamma/metabolism , Macrophages/immunology , Malaria/immunology , Malaria/mortality , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microbial Viability , Nitric Oxide/metabolism , Parasitemia/immunology , Plasmodium yoelii/immunology , Spleen/immunology , Spleen/metabolism , Vaccination , Vaccines, Attenuated/administration & dosage
10.
ACS Infect Dis ; 8(5): 998-1009, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35362944

ABSTRACT

Cerebral malaria (CM) is a serious central nervous system dysfunction caused by Plasmodium falciparum infection. In this study, we investigated the effect of Listeria monocytogenes (Lm) inoculation on experimental cerebral malaria (ECM) using Plasmodium berghei ANKA (PbA)-infected C57BL/6 mice. Live Lm inoculation inhibited the parasitemia and alleviated ECM symptoms. The protective effect against ECM symptoms was connected with improved brain pathology manifested as a less-damaged blood-brain barrier, decreased parasite sequestration, and milder local inflammation. Meanwhile, Lm inoculation decreased expression of cell adhesion molecules (ICAM-1 and VCAM-1) and accumulation of pathogenic CD8+ T cells in the brain. In keeping with the suppression of parasitemia, there was an upregulation of IFN-γ, IL-12, MCP-1, and NO expression in the spleen by Lm inoculation upon PbA infection. Early treatment with exogenous IFN-γ exhibited a similar effect to Lm inoculation on PbA infection. Taken together, Lm inoculation impedes the development of brain pathology in ECM, and early systemic IFN-γ production may play a critical role in these protective effects.


Subject(s)
Listeria monocytogenes , Malaria, Cerebral , Animals , Brain , CD8-Positive T-Lymphocytes/parasitology , CD8-Positive T-Lymphocytes/pathology , Malaria, Cerebral/parasitology , Malaria, Cerebral/pathology , Mice , Mice, Inbred C57BL , Parasitemia/pathology , Plasmodium berghei
11.
PLoS Genet ; 3(9): 1587-97, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17845078

ABSTRACT

Genetic makeup of the host plays a significant role in the course and outcome of infection. Inbred strains of mice display a wide range of sensitivities to Listeria monocytogenes infection and thus serve as a good model for analysis of the effect of genetic polymorphism. The outcome of L. monocytogenes infection in mice is influenced by the ability of this bacterium to induce expression of interferon beta mRNA, encoded in mouse by the Ifnb1 (interferon beta 1, fibroblast) gene. Mouse strains that lack components of the IFN beta signaling pathway are substantially more resistant to infection. We found that macrophages from the ByJ substrain of the common C57BL/6 inbred strain of mice are impaired in their ability to induce Ifnb1 expression in response to bacterial and viral infections. We mapped the locus that controls differential expression of Ifnb1 to a region on Chromosome 7 that includes interferon regulatory factor 3 (Irf3), which encodes a transcription factor responsible for early induction of Ifnb1 expression. In C57BL/6ByJ mice, Irf3 mRNA was inefficiently spliced, with a significant proportion of the transcripts retaining intron 5. Analysis of the Irf3 locus identified a single base-pair polymorphism and revealed that intron 5 of Irf3 is spliced by the atypical U12-type spliceosome. We found that the polymorphism disrupts a U12-type branchpoint and has a profound effect on the efficiency of splicing of Irf3. We demonstrate that a naturally occurring change in the splicing control element has a dramatic effect on the resistance to L. monocytogenes infection. Thus, the C57BL/6ByJ mouse strain serves as an example of how a mammalian host can counter bacterial virulence strategies by introducing subtle alteration of noncoding sequences.


Subject(s)
Interferon Regulatory Factor-3/genetics , Interferon-beta/biosynthesis , Listeriosis/metabolism , Polymorphism, Genetic , Animals , Cell Line , Interferon-beta/genetics , Listeriosis/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
12.
Int Immunopharmacol ; 64: 356-363, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30243072

ABSTRACT

Cerebral malaria (CM) is a serious and fatal malaria-associated syndrome caused by the development of an overwhelming proinflammatory response. Vitamin D (Vit.D; cholecalciferol) has regulatory functions associated with both innate and adaptive immune responses. Prevention is better than cure, in this experiment, we evaluated prophylactic oral Vit.D as a means of preventing CM presentation before infection of C57BL/6 mice with Plasmodium berghei ANKA (PbA) by modulating the host proinflammatory response. Mice that were supplemented with oral Vit.D has reduce death rate and ameliorated the integrity of the blood brain barrier. Prophylactic oral vitamin D relieved the symptoms of brain malaria and avoided death, gained valuable time for the diagnosis and treatment post infection. The robust Th1 response was attenuated in the Vit.D + PbA group. Furthermore, T-cell trafficking to the brain was diminished before PbA infection using Vit.D. The results suggest that Vit.D supplementation mediates the development of an anti-inflammatory environment that improves CM severity. In summary, the use of Vit.D as a nutritional supplement in malaria-endemic regions may help reduce the severity and mortality of CM.


Subject(s)
Malaria, Cerebral/prevention & control , Vitamin D/administration & dosage , Administration, Oral , Animals , Brain/immunology , Cell Adhesion/drug effects , Cell Movement , Dietary Supplements , Female , Interleukin-10/biosynthesis , Malaria, Cerebral/immunology , Mice , Mice, Inbred C57BL , Plasmodium berghei , Th1 Cells/immunology
13.
Parasit Vectors ; 9(1): 433, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27502144

ABSTRACT

BACKGROUND: Transmission-blocking vaccine (TBV) is a promising strategy for interrupting the malaria transmission cycle. Current TBV candidates include both pre- and post-fertilization antigens expressed during sexual development of the malaria parasites. METHODS: We tested whether a TBV design combining two sexual-stage antigens has better transmission-blocking activity. Using the rodent malaria model Plasmodium yoelii, we pursued a DNA vaccination strategy with genes encoding the gametocyte antigen Pys48/45 and the major ookinete surface protein Pys25. RESULTS: Immunization of mice with DNA constructs expression either Pys48/45 or Pys25 elicited strong antibody responses, which specifically recognized a ~45 and ~25 kDa protein from gametocyte and ookinete lysates, respectively. Immune sera from mice immunized with DNA constructs expressing Pys48/45 and Pys25 individually and in combination displayed evident transmission-blocking activity in in vitro ookinete culture and direct mosquito feeding experiments. With both assays, the Pys25 sera had higher transmission-blocking activity than the Pys48/45 sera. Intriguingly, compared with the immunization with the individual DNA vaccines, immunization with both DNA constructs produced lower antibody responses against individual antigens. The resultant immune sera from the composite vaccination had significantly lower transmission-blocking activity than those from Pys25 DNA immunization group, albeit the activity was substantially higher than that from the Pys48 DNA vaccination group. CONCLUSIONS: This result suggested that vaccination with the two DNA constructs did not achieve a synergistic effect, but rather caused interference in inducing antigen-specific antibody responses. This result has important implications for future design of composite vaccines targeting different sexual antigens.


Subject(s)
Antigens/immunology , Malaria Vaccines/immunology , Malaria/prevention & control , Plasmodium yoelii/immunology , Protozoan Proteins/immunology , Animals , Antigens/genetics , Disease Models, Animal , Female , Immunization , Malaria/immunology , Malaria/parasitology , Malaria/transmission , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Male , Mice , Mice, Inbred BALB C , Plasmodium yoelii/genetics , Plasmodium yoelii/physiology , Protozoan Proteins/administration & dosage , Protozoan Proteins/genetics , Reproduction
14.
Mol Med Rep ; 10(1): 353-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24737344

ABSTRACT

The present study aimed to investigate the signal transduction mechanism of the NZW type interleukin (IL)­10R1 and the effect on the function of B lymphocytes. Vectors for the expression of wild­type, NZW and G1146A IL­10R1 were constructed and transfected into Ba/F3 cells. The cell proliferation curve was determined using an MTT assay. CD32 and leukocyte-endothelial cell adhesion molecule 1 (LECAM­1) expression levels were assessed using flow cytometry. The phosphorylation of Janus kinase 1 (Jak1), tyrosine kinase 2 (Tyk2) and signal transducer and activator of transcription 3 (Stat3) were measured by western blot analysis. As compared with wild­type IL­10R1 cells, the proliferation ability of NZW IL­10R1 cells was increased following stimulation with IL­10, which was even greater for G1146A cells. CD32 levels in NZW IL­10R1 cells were lower, while the expression of LECAM­1 was higher compared with wild­type IL­10R1 cells, which was also observed for G1146A cells. The phosphorylation levels of Jak1 and Stat3 in NZW IL­10R1 and G1146A IL­10R1 cells were significantly lower compared with the wild­type IL­10R1 cells. However, the phosphorylation levels in the three cell types were not significantly different. In conclusion, the deficiency in Jak1­Stat3 signal transduction in NZW IL­10R1 cells induces a loss in the inhibition ability of proliferation and activation as well as a migration tendency of B lymphocytes, which is hypothesized to be associated with the occurrence and development of the autoimmune disease systemic lupus erythematosus (SLE).


Subject(s)
Interleukin-10 Receptor alpha Subunit/metabolism , Signal Transduction , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Line , Humans , Interleukin-10 Receptor alpha Subunit/genetics , Janus Kinase 1/metabolism , L-Selectin/metabolism , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Inbred C57BL , Mutagenesis , Phosphorylation , STAT3 Transcription Factor/metabolism , TYK2 Kinase/metabolism
15.
Parasitol Int ; 59(3): 400-6, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20609420

ABSTRACT

The outcome of Plasmodium yoelii 17XL (P.y17XL)-infected BALB/c and DBA/2 mice, ranging from death to spontaneous cure, depends largely on the establishment of effective Th1 and Th2 responses and a successful switch between Th1 and Th2 responses, as well as appropriate functioning of CD4(+)CD25(+)Foxp3(+)regulatory T cells (Tregs). The infection with another malaria-causing parasite, Plasmodium chabaudi AS (P.cAS), leads to a different outcome in BALB/c and DBA/2 mice compared to mice infected with P.y17XL alone. To understand the consequence of co-infection with P.y17XL and P.cAS, we determined the proliferation curve of parasites, pro-inflammatory/anti-inflammatory cytokine profiles, and the dynamic changes of the number of Tregs in DBA/2 and BALB/c mice with single or mixed-species infections. The infective mode in mixed-species infections was the same as single P.y17XL infections. The multiplication of P.y17XL parasites prevailed in BALB/c and DBA/2 mice with early mixed infections, as detected by RTQ-PCR. Subsequently, the multiplication of P.cAS parasites dominated in DBA/2 mice with mixed infections, while BALB/c mice succumbed to infection. In addition, the dynamic changes in IFN-gamma and IL-4 production in mice with mixed infections, used as a measure of Th1 and Th2 responsiveness, were consistent with P.y17XL-infected mice. Treg activation and the IL-10 level were also closely related to susceptibility to infection. Our findings demonstrate that the characteristics of the immune response during infections with mixed species are dependent on the mode of proliferation of different species of Plasmodium. Indeed, different species of Plasmodium can influence each other in the same host.


Subject(s)
Malaria/immunology , Malaria/parasitology , Plasmodium chabaudi/immunology , Plasmodium yoelii/immunology , Animals , Cytokines/metabolism , Female , Inflammation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Plasmodium chabaudi/pathogenicity , Plasmodium yoelii/pathogenicity , Species Specificity , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th2 Cells/immunology
16.
Immunogenetics ; 57(9): 697-702, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16160826

ABSTRACT

Several studies suggest that interleukin (IL)-10 pathway is involved in murine lupus, while no linkage of IL-10 gene polymorphism to disease susceptibility has been reported in studies with lupus-prone mice. Since IL-10 functions through the specific IL-10 receptor alpha (IL-10RA) chain and the IL-10RA gene (Il10ra) is linked to the susceptibility loci of atopic dermatitis and Crohn's disease identified using mouse models, we supposed that IL-10RA might be involved in murine lupus. By flow cytometry analysis, we found that NZW mice, one of the parental strains of lupus-prone (NZBxNZW) F1 mice, express extremely low levels of IL-10RA compared with NZB mice, the other parental strain, and the healthy BALB/c and C57BL/6 mice. Sequence analyses of Il10ra cDNA of NZW mice showed multiple nucleotide mutations compared with that of NZB and C57BL/6 strains, some of which would result in amino acid substitutions in the IL-10RA protein. Lupus-prone MRL mice shared the same polymorphism with NZW. Analyses using (NZBxNZW) F1xNZB backcross mice showed that high serum levels of IgG antichromatin antibodies were regulated by a combinatorial effect of the NZW Il10ra allele and a heterozygous genotype for Tnfa microsatellite locus. Our data suggest that the polymorphic NZW-type Il10ra may be involved in the pathologic production of antichromatin antibodies and, if so, may contribute in part to the development of systemic lupus erythematosus as one susceptibility allele.


Subject(s)
Genetic Predisposition to Disease , Lupus Erythematosus, Systemic/genetics , Polymorphism, Genetic , Receptors, Interleukin/genetics , Animals , Crosses, Genetic , DNA Primers , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Inbred NZB , Phenotype , Protein Subunits/genetics , Receptors, Interleukin/biosynthesis , Receptors, Interleukin-10 , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL