Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Front Immunol ; 10: 2673, 2019.
Article in English | MEDLINE | ID: mdl-31824484

ABSTRACT

It is known that Trypanosoma congolense infection in mice is associated with increased production of proinflammatory cytokines by macrophages and monocytes. However, the intracellular signaling pathways leading to the production of these cytokines still remain unknown. In this paper, we have investigated the innate receptors and intracellular signaling pathways that are associated with T. congolense-induced proinflammatory cytokine production in macrophages. We show that the production of IL-6, IL-12, and TNF-α by macrophages in vitro and in vivo following interaction with T. congolense is dependent on phosphorylation of mitogen-activated protein kinase (MAPK) including ERK, p38, JNK, and signal transducer and activation of transcription (STAT) proteins. Specific inhibition of MAPKs and STATs signaling pathways significantly inhibited T. congolense-induced production of proinflammatory cytokines in macrophages. We further show that T. congolense-induced proinflammatory cytokine production in macrophages is mediated via Toll-like receptor 2 (TLR2) and involves the adaptor molecule, MyD88. Deficiency of MyD88 and TLR2 leads to impaired cytokine production by macrophages in vitro and acute death of T. congolense-infected relatively resistant mice. Collectively, our results provide insight into T. congolense-induced activation of the immune system that leads to the production of proinflammatory cytokines and resistance to the infection.


Subject(s)
Myeloid Differentiation Factor 88/metabolism , Toll-Like Receptor 2/metabolism , Trypanosomiasis, African/immunology , Trypanosomiasis, African/metabolism , Adenylate Kinase/immunology , Adenylate Kinase/metabolism , Animals , Cytokines/biosynthesis , Enzyme Activation/immunology , Female , Macrophages/immunology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/immunology , STAT Transcription Factors/immunology , STAT Transcription Factors/metabolism , Toll-Like Receptor 2/immunology , Trypanosoma congolense/immunology
2.
J Immunol ; 203(4): 964-971, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31243088

ABSTRACT

NK cells are key innate immune cells that play critical roles in host defense. Although NK cells have been shown to regulate immunity to some infectious diseases, their role in immunity to Trypanosoma congolense has not been investigated. NK cells are vital sources of IFN-γ and TNF-α; two key cytokines that are known to play important roles in resistance to African trypanosomes. In this article, we show that infection with T. congolense leads to increased levels of activated and functional NK cells in multiple tissue compartments. Systemic depletion of NK cells with anti-NK1.1 mAb led to increased parasitemia, which was accompanied by significant reduction in IFN-γ production by immune cells in the spleens and liver of infected mice. Strikingly, infected NFIL3-/- mice (which genetically lack NK cell development and function) on the normally resistant background were highly susceptible to T. congolense infection. These mice developed fulminating and uncontrolled parasitemia and died significantly earlier (13 ± 1 d) than their wild-type control mice (106 ± 26 d). The enhanced susceptibility of NFIL3-/- mice to infection was accompanied by significantly impaired cytokine (IFN-γ and TNF-α) response by CD3+ T cells in the spleens and liver. Adoptive transfer of NK cells into NFIL3-/- mice before infection rescued them from acute death in a perforin-dependent manner. Collectively, these studies show that NK cells are critical for optimal resistance to T. congolense, and its deficiency leads to enhanced susceptibility in infected mice.


Subject(s)
Killer Cells, Natural/immunology , Trypanosomiasis, African/immunology , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Trypanosoma congolense/immunology
3.
Immunol Res ; 67(1): 84-92, 2019 02.
Article in English | MEDLINE | ID: mdl-30467677

ABSTRACT

Diminazene aceturate (Berenil) is the most commonly used trypanolytic agent in livestock. We previously showed that Berenil downregulates Trypanosoma congolense (T. congolense)-induced cytokine production in macrophages both in vitro and in vivo. Here, we investigated the molecular mechanisms through which the drug alters T. congolense-induced cytokine production in macrophages. We show that pretreatment of macrophages with Berenil significantly downregulated T. congolense-induced phosphorylation of mitogen-activated protein kinase (p38), signal transducer and activator of transcription (STAT) proteins including STAT1 and STAT3, and NFκB activity both in vitro and in vivo. Collectively, our results reveal a mechanistic insight through which Berenil downregulates T. congolense-induced cytokine production in macrophages by inhibiting key signaling molecules and pathways associated with proinflammatory cytokine production.


Subject(s)
Diminazene/analogs & derivatives , Macrophages/immunology , Trypanocidal Agents/therapeutic use , Trypanosoma congolense/physiology , Trypanosomiasis, African/drug therapy , Animals , Cattle , Cell Line, Transformed , Cytokines/metabolism , Diminazene/therapeutic use , Female , Humans , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphorylation , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Trypanosomiasis, African/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Sci Rep ; 8(1): 12969, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30154572

ABSTRACT

Breast cancer is the most common cancer in women worldwide. Hormone receptor breast cancers are the most common ones and, about 2 out of every 3 cases of breast cancer are estrogen receptor (ER) positive. Selective ER modulators, such as tamoxifen, are the first line of endocrine treatment of breast cancer. Despite the expression of hormone receptors some patients develop tamoxifen resistance and 50% present de novo tamoxifen resistance. Recently, we have demonstrated that activated mammalian target of rapamycin (mTOR) is positively associated with overall survival and recurrence free survival in ER positive breast cancer patients who were later treated with tamoxifen. Since altered expression of protein kinase B (PKB)/Akt in breast cancer cells affect N-myristoyltransferase 1 (NMT1) expression and activity, we investigated whether mTOR, a downstream target of PKB/Akt, regulates NMT1 in ER positive breast cancer cells (MCF7 cells). We inhibited mTOR by treating MCF7 cells with rapamycin and observed that the expression of NMT1 increased with rapamycin treatment over the period of time with a concomitant decrease in mTOR phosphorylation. We further employed mathematical modelling to investigate hitherto not known relationship of mTOR with NMT1. We report here for the first time a collection of models and data validating regulation of NMT1 by mTOR.


Subject(s)
Acyltransferases/biosynthesis , Adenocarcinoma/enzymology , Breast Neoplasms/enzymology , Estrogens , Neoplasm Proteins/physiology , Neoplasms, Hormone-Dependent/enzymology , TOR Serine-Threonine Kinases/physiology , Acyltransferases/genetics , Enzyme Induction , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Models, Biological , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Estrogen/analysis , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors
5.
J Immunol ; 201(2): 507-515, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29898961

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of bone marrow-derived myeloid cells that have immune-suppressive activities. These cells have been reported to suppress T cell immunity against tumors as well as in some parasitic and bacterial infections. However, their role during Trypanosoma congolense infection has not been studied. Given that immunosuppression is a hallmark of African trypanosomiasis, we investigated the role of MDSCs in immunity to T. congolense infection. We found increased numbers of MDSCs in the spleen and liver of infected mice, which correlated with increased parasitemia. Depletion of MDSCs significantly increased the percentage of proliferating and IFN-γ-producing CD4+ T cells from the spleen of T. congolense-infected mice. Furthermore, MDSCs from T. congolense-infected mice directly suppressed CD4+ T cell proliferation in a coculture setting. This suppressive effect was abolished by the arginase-1 inhibitor, Nω-hydroxy-nor-l-arginine (nor-NOHA), indicating that MDSCs suppress CD4+ T cell proliferation and function in an arginase-1-dependent manner. Indeed, depletion of MDSCs during infection led to control of the first wave of parasitemia and prolonged survival of infected mice. This was also associated with increased CD4+ T cell proliferation and IFN-γ production. Taken together, our findings identify an important role of MDSCs in the pathogenesis of experimental T. congolense infection via suppression of T cell proliferative and effector cytokine responses in an arginase-1-dependent manner.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Proliferation/physiology , Interferon-gamma/immunology , Myeloid-Derived Suppressor Cells/immunology , Trypanosoma congolense/immunology , Trypanosomiasis, African/immunology , Animals , Arginase/immunology , Female , Immune Tolerance/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Myeloid Cells/immunology , Spleen/immunology
6.
Front Immunol ; 7: 181, 2016.
Article in English | MEDLINE | ID: mdl-27242788

ABSTRACT

Pathogens, such as bacteria, viruses, and parasites, possess specific molecules or proteins that are recognized by several host innate immune receptors, leading to the activation of several intracellular signaling molecules and pathways. The magnitude and quality of these events significantly affect the outcome of infection. African trypanosomes, including Trypanosoma congolense, are capable of manipulating the host immune response, including the activity of macrophages, which are the key immune cells that contribute to the immunopathogenesis of African trypanosomiasis. Although it is known that immune hyperactivation and excessive pro-inflammatory cytokine production are the hallmarks of African trypanosomiasis, the mechanisms through which these events are triggered are poorly defined. However, it is known that macrophages may play a significant role in these processes, because phagocytosis of trypanosomes by macrophages initiates intracellular signal transduction cascades that lead to the release of pro-inflammatory cytokines and alteration in cell function. This review highlights recent progress in our understanding of the innate immune receptors, signaling pathways, and transcription factors involved in T. congolense-induced pro-inflammatory cytokine production in macrophages. It will reveal the existence of complex signaling events through which the parasite modulates the host immune response, thus identifying novel targets that could aid in designing strategies to effectively control the disease.

7.
PLoS Negl Trop Dis ; 9(4): e0003716, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25875604

ABSTRACT

BACKGROUND: Bam32, a 32 kDa adaptor molecule, plays important role in B cell receptor signalling, T cell receptor signalling and antibody affinity maturation in germinal centres. Since antibodies against trypanosome variant surface glycoproteins (VSG) are critically important for control of parasitemia, we hypothesized that Bam32 deficient (Bam32-/-) mice would be susceptible to T. congolense infection. METHODOLOGY/PRINCIPAL FINDINGS: We found that T. congolense-infected Bam32-/- mice successfully control the first wave of parasitemia but then fail to control subsequent waves and ultimately succumb to their infection unlike wild type (WT) C57BL6 mice which are relatively resistant. Although infected Bam32-/- mice had significantly higher hepatomegaly and splenomegaly, their serum AST and ALT levels were not different, suggesting that increased liver pathology may not be responsible for the increased susceptibility of Bam32-/- mice to T. congolense. Using direct ex vivo flow cytometry and ELISA, we show that CD4+ T cells from infected Bam32-/- mice produced significantly increased amounts of disease-exacerbating proinflammatory cytokines (including IFN-γ, TNF-α and IL-6). However, the percentages of regulatory T cells and IL-10-producing CD4+ cells were similar in infected WT and Bam32-/- mice. While serum levels of parasite-specific IgM antibodies were normal, the levels of parasite-specific IgG, (particularly IgG1 and IgG2a) were significantly lower in Bam32-/- mice throughout infection. This was associated with impaired germinal centre response in Bam32-/- mice despite increased numbers of T follicular helper (Tfh) cells. Adoptive transfer studies indicate that intrinsic B cell defect was responsible for the enhanced susceptibility of Bam32-/- mice to T. congolense infection. CONCLUSIONS/SIGNIFICANCE: Collectively, our data show that Bam32 is important for optimal anti-trypanosome IgG antibody response and suppression of disease-promoting proinflammatory cytokines and its deficiency leads to inability to control T. congolense infection in mice.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Lipoproteins/metabolism , Trypanosoma congolense/immunology , Trypanosomiasis, African/immunology , Adaptor Proteins, Signal Transducing/genetics , Adoptive Transfer , Animals , Antibody Affinity , Antibody Formation , B-Lymphocytes/immunology , Cytokines/metabolism , Disease Susceptibility , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Lipoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Parasitemia/immunology , T-Lymphocytes, Regulatory , Trypanosomiasis, African/parasitology
8.
Int Immunopharmacol ; 21(2): 342-5, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24893117

ABSTRACT

Diminazene aceturate or Berenil has been the drug of choice for treatment of animal trypanosomiasis. Although the compound has been in the market since 1955, its mechanisms of action have remained poorly understood. While some earlier reports show that Berenil possesses trypanolytic and trypanostatic properties, some studies show it may also indirectly affect the host immune system. Our recent extensive studies show that treatment with Berenil reduces pro-inflammatory cytokine (IL-6, IL-12 and TNF) production in macrophages in vivo and in vitro following stimulation with Trypanosoma congolense, lipopolysaccharide (LPS), unmethylated bacterial CpG motifs and Poly I:C. This global effect was not due to downregulation of Toll-like receptor (TLR) expression on innate immune cells. Instead, Berenil significantly downregulated phosphorylation of mitogen activated protein kinases (MAPKs, including ERK, p38 and JNK), signal transducer and activator of transcription (STAT) proteins (including STAT1 and STAT3) and NFκB p65 subunit, key signaling molecules and transcription factors involved in the production of proinflammatory cytokines. The ability of Berenil to downregulate major intracellular signaling pathways that lead to proinflammatory cytokine production suggests that it could be used to treat conditions caused by excessive production of inflammatory cytokines.


Subject(s)
Diminazene/analogs & derivatives , Signal Transduction/drug effects , Animals , Cytokines/metabolism , Diminazene/pharmacology , Diminazene/therapeutic use , Humans , Inflammation/drug therapy , Inflammation/metabolism , Transcription Factors/metabolism
9.
Innate Immun ; 20(7): 760-73, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24179040

ABSTRACT

Although diminazene aceturate (Berenil) is widely used as a trypanolytic agent in livestock, its mechanisms of action remain poorly understood. We previously showed that Berenil treatment suppresses pro-inflammatory cytokine production by splenic and liver macrophages leading to a concomitant reduction in serum cytokine levels in mice infected with Trypanosoma congolense or challenged with LPS. Here, we investigated the molecular mechanisms through which Berenil alters pro-inflammatory cytokine production by macrophages. We show that pre-treatment of macrophages with Berenil dramatically suppressed IL-6, IL-12 and TNF-α production following LPS, CpG and Poly I:C stimulation without altering the expression of TLRs. Instead, it significantly down-regulated phosphorylation of mitogen-activated protein kinases (p38, extracellular signal-regulated kinase and c-Jun N-terminal kinases), signal transducer and activator of transcription (STAT) proteins (STAT1 and STAT3) and NF-кB p65 activity both in vitro and in vivo. Interestingly, Berenil treatment up-regulated the phosphorylation of STAT5 and the expression of suppressor of cytokine signaling 1 (SOCS1) and SOCS3, which are negative regulators of innate immune responses, including MAPKs and STATs. Collectively, these results show that Berenil down-regulates macrophage pro-inflammatory cytokine production by inhibiting key signaling pathways associated with cytokine production and suggest that this drug may be used to treat conditions caused by excessive production of inflammatory cytokines.


Subject(s)
Cytokines/biosynthesis , Diminazene/analogs & derivatives , Inflammation/chemically induced , Inflammation/metabolism , Lipopolysaccharides/antagonists & inhibitors , Mitogen-Activated Protein Kinases/antagonists & inhibitors , STAT Transcription Factors/antagonists & inhibitors , Trypanocidal Agents/toxicity , Animals , Diminazene/toxicity , Down-Regulation/drug effects , Female , Interleukin-6/biosynthesis , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Phosphorylation/drug effects , STAT Transcription Factors/metabolism , Suppressor of Cytokine Signaling Proteins/antagonists & inhibitors , Toll-Like Receptors/biosynthesis
10.
Infect Immun ; 82(3): 1074-83, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24343657

ABSTRACT

BALB/c mice are highly susceptible to experimental intraperitoneal Trypanosoma congolense infection. However, a recent report showed that these mice are relatively resistant to primary intradermal low-dose infection. Paradoxically, repeated low-dose intradermal infections predispose mice to enhanced susceptibility to an otherwise noninfectious dose challenge. Here, we explored the mechanisms responsible for this low-dose-induced susceptibility to subsequent low-dose challenge infection. We found that akin to intraperitoneal infection, low-dose intradermal infection led to production of interleukin-10 (IL-10), IL-6, IL-12, tumor necrosis factor alpha (TNF-α), transforming growth factor ß (TGF-ß), and gamma interferon (IFN-γ) by spleen and draining lymph node cells. Interestingly, despite the absence of parasitemia, low-dose intradermal infection led to expansion of CD4+ CD25+ Foxp3+ cells (T regulatory cells [Tregs]) in both the spleens and lymph nodes draining the infection site. Depletion of Tregs by anti-CD25 monoclonal antibody (MAb) treatment during primary infection or before challenge infection following repeated low-dose infection completely abolished the low-dose-induced enhanced susceptibility. In addition, Treg depletion was associated with dramatic reduction in serum levels of TGF-ß and IL-10. Collectively, these findings show that low-dose intradermal infection leads to rapid expansion of Tregs, and these cells mediate enhanced susceptibility to subsequent infection.


Subject(s)
Disease Susceptibility/immunology , T-Lymphocytes, Regulatory/immunology , Trypanosoma congolense/immunology , Trypanosomiasis, African/immunology , Animals , Antibodies, Monoclonal/immunology , CD4 Antigens/immunology , Cells, Cultured , Disease Susceptibility/parasitology , Female , Forkhead Transcription Factors/immunology , Interferon-gamma/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Interleukins/immunology , Lymph Nodes/immunology , Lymph Nodes/parasitology , Mice , Mice, Inbred BALB C , Parasitemia/immunology , Parasitemia/parasitology , Spleen/immunology , Spleen/parasitology , T-Lymphocytes, Regulatory/parasitology , Transforming Growth Factor beta/immunology , Trypanosomiasis, African/parasitology , Tumor Necrosis Factor-alpha/immunology
11.
J Immunol ; 190(7): 3380-9, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23460745

ABSTRACT

The breakdown of L-arginine to ornithine and urea by host arginase supports Leishmania proliferation in macrophages. Studies using arginase-null mutants show that Leishmania-derived arginase plays an important role in disease pathogenesis. We investigated the role of parasite-derived arginase in secondary (memory) anti-Leishmania immunity in the resistant C57BL/6 mice. We found that C57BL/6 mice infected with arginase-deficient (arg(-)) L. major failed to completely resolve their lesion and maintained chronic pathology after 16 wk, a time when the lesion induced by wild-type L. major is completely resolved. This chronic disease was associated with impaired Ag-specific proliferation and IFN-γ production, a concomitant increase in programmed cell death-1 (PD-1) expression on CD4(+) T cells, and failure to induce protection against secondary L. major challenge. Treatment with anti-PD-1 mAb restored T cell proliferation and IFN-γ production in vitro and led to complete resolution of chronic lesion in arg(-) L. major-infected mice. These results show that infection with arg(-) L. major results in chronic disease due in part to PD-1-mediated clonal exhaustion of T cells, suggesting that parasite-derived arginase contributes to the overall quality of the host immune response and subsequent disease outcome in L. major-infected mice. They also indicate that persistent parasites alone do not regulate the quality of secondary anti-Leishmania immunity in mice and that the quality of the primary immune response may be playing a hitherto unrecognized dominant role in this process.


Subject(s)
Arginase/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Leishmania major/enzymology , Leishmania major/immunology , Programmed Cell Death 1 Receptor/metabolism , Animals , Arginase/genetics , Cytokines/immunology , Cytokines/metabolism , Female , Immunologic Memory , Leishmania major/genetics , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/metabolism , Mice , Mice, Inbred C57BL
12.
PLoS One ; 7(11): e48696, 2012.
Article in English | MEDLINE | ID: mdl-23144931

ABSTRACT

BACKGROUND: Trypanosoma congolense are extracellular and intravascular blood parasites that cause debilitating acute or chronic disease in cattle and other domestic animals. Diminazene aceturate (Berenil) has been widely used as a chemotherapeutic agent for trypanosomiasis in livestock since 1955. As in livestock, treatment of infected highly susceptible BALB/c mice with Berenil leads to rapid control of parasitemia and survival from an otherwise lethal infection. The molecular and biochemical mechanisms of action of Berenil are still not very well defined and its effect on the host immune system has remained relatively unstudied. Here, we investigated whether Berenil has, in addition to its trypanolytic effect, a modulatory effect on the host immune response to Trypanosoma congolense. METHODOLOGY/PRINCIPAL FINDINGS: BALB/c and C57BL/6 mice were infected intraperitoneally with T. congolense, treated with Berenil and the expression of CD25 and FoxP3 on splenic cells was assessed directly ex vivo. In addition, serum levels and spontaneous and LPS-induced production of pro-inflammatory cytokines by splenic and hepatic CD11b⁺ cells were determined by ELISA. Berenil treatment significantly reduced the percentages of CD25⁺ cells, a concomitant reduction in the percentage of regulatory (CD4⁺Foxp3⁺) T cells and a striking reduction in serum levels of disease exacerbating pro-inflammatory cytokines including IL-6, IL-12, TNF and IFN-γ. Furthermore, Berenil treatment significantly suppressed spontaneous and LPS-induced production of inflammatory cytokines by splenic and liver macrophages and significantly ameliorated LPS-induced septic shock and the associated cytokine storm. CONCLUSIONS/SIGNIFICANCE: Collectively, these results provide evidence that in addition to its direct trypanolytic effect, Berenil also modulates the host immune response to the parasite in a manner that dampen excessive immune activation and production of pathology-promoting pro-inflammatory cytokines, suggesting that this drug may also be beneficial for treatment of disease conditions caused by excessive production of inflammatory cytokines.


Subject(s)
Diminazene/analogs & derivatives , Immunity, Cellular/drug effects , Inflammation/drug therapy , Trypanosoma congolense , Trypanosomiasis, African/drug therapy , Animals , Cytokines/blood , Cytokines/metabolism , Diminazene/pharmacology , Diminazene/therapeutic use , Enzyme-Linked Immunosorbent Assay , Female , Forkhead Transcription Factors/metabolism , Inflammation/etiology , Interleukin-2 Receptor alpha Subunit/metabolism , Kupffer Cells/immunology , Kupffer Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Spleen/cytology , Spleen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Trypanosomiasis, African/complications
13.
PLoS Negl Trop Dis ; 6(7): e1761, 2012.
Article in English | MEDLINE | ID: mdl-22860150

ABSTRACT

BACKGROUND: BALB/c mice are highly susceptible while C57BL/6 are relatively resistant to experimental Trypanosoma congolense infection. Although regulatory T cells (Tregs) have been shown to regulate the pathogenesis of experimental T. congolense infection, their exact role remains controversial. We wished to determine whether Tregs contribute to distinct phenotypic outcomes in BALB/c and C57BL/6 mice and if so how they operate with respect to control of parasitemia and production of disease-exacerbating proinflammatory cytokines. METHODOLOGY/FINDINGS: BALB/c and C57BL/6 mice were infected intraperitoneally (i.p) with 10(3)T. congolense clone TC13 and both the kinetics of Tregs expansion and intracellular cytokine profiles in the spleens and livers were monitored directly ex vivo by flow cytometry. In some experiments, mice were injected with anti-CD25 mAb prior or post T. congolense infection or adoptively (by intravenous route) given highly enriched naïve CD25(+) T lymphocytes prior to T. congolense infection and the inflammatory cytokine/chemokine levels and survival were monitored. In contrast to a transient and non significant increase in the percentages and absolute numbers of CD4(+)CD25(+)Foxp3(+) T cells (Tregs) in C57BL/6 mouse spleens and livers, a significant increase in the percentage and absolute numbers of Tregs was observed in spleens of infected BALB/c mice. Ablation or increasing the number of CD25(+) cells in the relatively resistant C57BL/6 mice by anti-CD25 mAb treatment or by adoptive transfer of CD25(+) T cells, respectively, ameliorates or exacerbates parasitemia and production of proinflammatory cytokines. CONCLUSION: Collectively, our results show that regulatory T cells contribute to susceptibility in experimental murine trypanosomiasis in both the highly susceptible BALB/c and relatively resistant C57BL/6 mice.


Subject(s)
T-Lymphocytes, Regulatory/immunology , Trypanosoma congolense/immunology , Trypanosoma congolense/pathogenicity , Trypanosomiasis, African/immunology , Trypanosomiasis, African/pathology , Animals , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Female , Flow Cytometry , Liver/immunology , Liver/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Parasitemia/immunology , Spleen/immunology , Spleen/parasitology
14.
Infect Immun ; 78(12): 5271-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20855512

ABSTRACT

BALB/c mice are highly susceptible to experimental Trypanosoma congolense infections, whereas C57BL/6 mice are relatively resistant. Infected highly susceptible BALB/c mice die of systemic inflammatory response syndrome. Because interleukin-17 (IL-17) and Th17 cells regulate inflammatory responses, we investigated their role in the pathogenesis of experimental African trypanosomiasis in mice. We show that the production of IL-17 by spleen and liver cells and the serum IL-17 level increased after T. congolense infection in mice. Interestingly, infected highly susceptible BALB/c mice produced more IL-17 and had more Th17 cells than infected relatively resistant C57BL/6 mice. Paradoxically, neutralization of IL-17 with anti-IL-17 monoclonal antibody in vivo induced higher parasitemia in both the susceptible and the relatively resistant mice. Interestingly, anti-IL-17 antibody-treated mice had higher serum levels of alanine aminotransferase and aspartate aminotransferase, and the production of IL-10 and nitric oxide by liver cells was markedly decreased. Moreover, recombinant IL-17-treated mice exhibited significantly faster parasite control and lower peak parasitemia compared to control mice. Collectively, these results suggest that the IL-17/Th17 axis plays a protective role in murine experimental African trypanosomiasis.


Subject(s)
Interleukin-17/physiology , Parasitemia/immunology , Trypanosoma congolense/immunology , Trypanosomiasis, African/immunology , Animals , Antibodies, Protozoan/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Immunity, Innate/immunology , Interleukin-17/blood , Mice , Mice, Inbred BALB C/immunology , Mice, Inbred BALB C/parasitology , Mice, Inbred C57BL/immunology , Mice, Inbred C57BL/parasitology , Parasitemia/parasitology , Trypanosomiasis, African/physiopathology
15.
Vaccine ; 28(33): 5451-7, 2010 Jul 26.
Article in English | MEDLINE | ID: mdl-20558242

ABSTRACT

It is widely believed that persistence of live parasites at the primary site of infection is important for maintenance of anti-Leishmania immunity. However, whether this immunity requires only the presence of antigen and not necessarily live replicating parasites has not been investigated. To determine whether non-replicating antigens could induce and maintain anti-Leishmania immunity, we inoculated naïve mice with killed parasites (once or 5 times weekly) either alone or in combination with rIL-12 and challenged them with virulent Leishmania major parasites at different times after inoculation. We found that similar to mice that recovered from virulent live L. major infection, mice inoculated repeatedly with killed parasites were protected against virulent L. major challenge. The protection obtained following 5 weekly inoculations of killed parasites was associated with strong antigen-specific IFN-gamma production by cells from the lymph nodes draining the inoculation site. In contrast, mice that received a single or double inoculation of killed parasites either alone or followed with repeated rIL-12 injection were not protected. Repeated antigen inoculation resulted in increased numbers of the IFN-gamma-secreting CD44(+)CD62L(-) T cells that were comparable in magnitude to that seen in mice with persistent infections. Overall, these results suggest that it is possible to generate and maintain anti-Leishmania immunity for a relatively long period of time in the absence of live replicating parasites. However, a certain threshold of effector cells has to be generated in order to achieve this protection.


Subject(s)
Interleukin-12/pharmacology , Leishmania major/immunology , Leishmaniasis Vaccines/pharmacology , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/prevention & control , Recombinant Proteins/pharmacology , Animals , Interferon-gamma/immunology , Interleukin-12/immunology , Leishmaniasis Vaccines/immunology , Mice , Recombinant Proteins/immunology , T-Lymphocytes/immunology , Time Factors , Vaccines, Inactivated/immunology , Vaccines, Inactivated/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...