Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 211
Filter
Add more filters

Publication year range
1.
Annu Rev Cell Dev Biol ; 35: 381-406, 2019 10 06.
Article in English | MEDLINE | ID: mdl-31283378

ABSTRACT

Innate immunity and adaptive immunity consist of highly specialized immune lineages that depend on transcription factors for both function and development. In this review, we dissect the similarities between two innate lineages, innate lymphoid cells (ILCs) and dendritic cells (DCs), and an adaptive immune lineage, T cells. ILCs, DCs, and T cells make up four functional immune modules and interact in concert to produce a specified immune response. These three immune lineages also share transcriptional networks governing the development of each lineage, and we discuss the similarities between ILCs and DCs in this review.


Subject(s)
Adaptive Immunity , Dendritic Cells/immunology , Gene Regulatory Networks , Immunity, Innate/genetics , Lymphocytes/immunology , Animals , Cell Differentiation/immunology , Cytokines/metabolism , Gene Expression Regulation/immunology , Humans , T-Lymphocytes/immunology , T-Lymphocytes/microbiology , T-Lymphocytes/parasitology , T-Lymphocytes/virology , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Immunol Rev ; 292(1): 139-148, 2019 11.
Article in English | MEDLINE | ID: mdl-31553065

ABSTRACT

T cells exposed to persistent antigen in the inflammatory environment of chronic infections often show progressive loss of effector functions, high expression of inhibitory receptors and distinct transcriptional programs. T cells in this functional state are termed "exhausted" and T cell exhaustion is associated with inefficient control of infections. A remarkably similar scenario has been described for B cells during chronic infections in humans, including malaria, in which case a subpopulation of atypical memory B cells (MBCs) greatly expands and these MBCs show attenuation of B cell receptor signaling, loss of the B cell effector functions of antibody and cytokine production, high expression of inhibitory receptors and distinct transcriptional profiles. The expansion of these MBCs is also associated with inefficient control of infections. Despite the similarities with exhausted T cells we speculate that at least in malaria, atypical MBCs may not be exhausted but rather may be functional, possibly even beneficial. Our recent results suggest that we simply may not have known how to ask an atypical MBC to function. Thus, exhaustion may not be in the human B cell's vocabulary, at least not in malaria.


Subject(s)
Antigens, Protozoan/immunology , B-Lymphocytes/immunology , Immunologic Memory/immunology , Malaria/immunology , T-Lymphocytes/immunology , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/parasitology , Humans , Malaria/parasitology , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, B-Cell/metabolism , Signal Transduction/immunology , T-Lymphocytes/parasitology
3.
Cytokine ; 145: 155208, 2021 09.
Article in English | MEDLINE | ID: mdl-32736961

ABSTRACT

Dendritic cells (DCs), as antigen-presenting cells, can reportedly be infected withLeishmaniaparasites and hence provide a better option to trigger T-cell primary immune responses and immunological memory. We consistently primed DCs during culture with purified recombinant cytosolic tryparedoxin (rcTXN) and then evaluated the vaccine prospect of presentation of rcTXN against VL in BALB/c mice. We reported earlier the immunogenic properties of cTXN antigen derived fromL. donovani when anti-cTXN antibody was detected in the sera of kala-azar patients. It was observed that cTXN antigen, when used as an immunogen with murine DCs acting as a vehicle, was able to induce complete protection against VL in an infected group of immunized mice. This vaccination triggered splenic macrophages to produce more IL-12 and GM-CSF, and restricted IL-10 release to a minimum in an immunized group of infected animals. Concomitant changes in T-cell responses against cTXN antigen were also noticed, which increased the release of protective cytokine-like IFN-γ under the influence of NF-κß in the indicated vaccinated group of animals. All cTXN-DCs-vaccinated BALB/c mice survived during the experimental period of 120 days. The results obtained in our study suggest that DCs primed with cTXN can be used as a vaccine prospect for the control of visceral leishmaniasis.


Subject(s)
Dendritic Cells/immunology , Leishmania donovani/immunology , Leishmaniasis Vaccines/immunology , Leishmaniasis, Visceral/immunology , Animals , Cytokines/immunology , Dendritic Cells/parasitology , Immunity, Cellular/immunology , Interleukin-10/immunology , Interleukin-12/immunology , Leishmaniasis, Visceral/parasitology , Macrophages/immunology , Macrophages/parasitology , Mice , Mice, Inbred BALB C , Protozoan Proteins/immunology , T-Lymphocytes/immunology , T-Lymphocytes/parasitology
4.
Cell Microbiol ; 21(7): e13021, 2019 07.
Article in English | MEDLINE | ID: mdl-30835870

ABSTRACT

Protozoan pathogens secrete nanosized particles called extracellular vesicles (EVs) to facilitate their survival and chronic infection. Here, we show the inhibition by Plasmodium berghei NK65 blood stage-derived EVs of the proliferative response of CD4+ T cells in response to antigen presentation. Importantly, these results were confirmed in vivo by the capacity of EVs to diminish the ovalbumin-specific delayed type hypersensitivity response. We identified two proteins associated with EVs, the histamine releasing factor (HRF) and the elongation factor 1α (EF-1α) that were found to have immunosuppressive activities. Interestingly, in contrast to WT parasites, EVs from genetically HRF- and EF-1α-deficient parasites failed to inhibit T cell responses in vitro and in vivo. At the level of T cells, we demonstrated that EVs from WT parasites dephosphorylate key molecules (PLCγ1, Akt, and ERK) of the T cell receptor signalling cascade. Remarkably, immunisation with EF-1α alone or in combination with HRF conferred a long-lasting antiparasite protection and immune memory. In conclusion, we identified a new mechanism by which P. berghei-derived EVs exert their immunosuppressive functions by altering T cell responses. The identification of two highly conserved immune suppressive factors offers new conceptual strategies to overcome EV-mediated immune suppression in malaria-infected individuals.


Subject(s)
Biomarkers, Tumor/genetics , Extracellular Vesicles/immunology , Malaria/genetics , Peptide Elongation Factor 1/genetics , Animals , Antigen Presentation/immunology , Antigens/genetics , Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/parasitology , Cell Proliferation/genetics , Extracellular Vesicles/genetics , Humans , Immune Evasion/genetics , Immune Evasion/immunology , Malaria/parasitology , Malaria/pathology , Plasmodium berghei/genetics , Plasmodium berghei/pathogenicity , T-Lymphocytes/immunology , T-Lymphocytes/parasitology , Tumor Protein, Translationally-Controlled 1
5.
Biologicals ; 67: 42-48, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32718776

ABSTRACT

Plasmodium falciparum leads to a virulent form of malaria. Progress has been achieved in understanding the mechanisms involved in the malarial infection, still there is no effective vaccine to prevent severe infection. An effective vaccine against malaria should be one which can induce immune responses against multiple epitopes in the context of predominantly occurring HLA alleles. In this study, an integrated approach was employed to identify promiscuous peptides of a well-defined sequence of erythrocyte binding antigen-175 and promiscuous peptides for HLA alleles were designed using bioinformatics tools. A peptide with 15 amino acids (ILAIAIYESRILKRK) was selected based on its high binding affinity score and synthesized. This promiscuous peptide was used as stimulating antigen in lymphoproliferative responses to evaluate the cellular immune response. It was observed this peptide evokes lymphoproliferative and cytokine responses in individuals naturally exposed to the malaria parasite. The intensity of PBMCs proliferation was observed to be higher in sera obtained from P. falciparum exposed as compared to unexposed healthy individuals, suggesting earlier recognition of peptide of this region by T cells. Furthermore, the binding mode of HLA-peptide complex and their interaction may lead to a rational and selective peptide-based vaccine candidate design approach which can be used as a malaria prophylaxis.


Subject(s)
Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Peptides/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Alleles , Amino Acid Sequence , Antigens, Protozoan/metabolism , Cells, Cultured , Drug Design , HLA Antigens/genetics , HLA Antigens/immunology , HLA Antigens/metabolism , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , Malaria Vaccines/administration & dosage , Malaria, Falciparum/parasitology , Malaria, Falciparum/prevention & control , Molecular Docking Simulation , Peptides/chemistry , Peptides/metabolism , Plasmodium falciparum/metabolism , Plasmodium falciparum/physiology , Protein Binding , Protozoan Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/parasitology
6.
Vet Pathol ; 57(2): 272-280, 2020 03.
Article in English | MEDLINE | ID: mdl-31801417

ABSTRACT

Sarcocystosis is a protozoal disease affecting a wide range of animals. The aims of this study were to characterize the following in sheep: (1) the muscle pathology in Sarcocystis infection, (2) the inflammatory infiltrate and its relationship to severity of infection, and (3) immune markers expressed by parasitized muscle fibers and parasitic cysts. Skeletal muscle samples from 78 sheep slaughtered in southern Italy were snap frozen and analyzed by histopathology, immunohistochemistry, and immunofluorescence. Polymerase chain reaction (PCR) and sequencing were used for Sarcocystis species identification. All 40 muscle samples tested were PCR-positive for Sarcocystis tenella. Histologically, cysts were identified in 76/78 cases (97%), associated with an endomysial infiltrate of lymphocytes and plasma cells. The T cells were predominantly CD8+, with fewer CD4+ or CD79α+ cells. Eosinophils were absent. Notably, sarcolemmal immunopositivity for major histocompatibility complex (MHC) I and II was found in 76/78 cases (97%) and 75/78 cases (96%), respectively, both in samples with and in those without evident inflammatory infiltrate. The number of cysts was positively correlated with inflammation. In addition, MHC I was detected in 55/78 cyst walls (72%), and occasionally co-localized with the membrane-associated protein dystrophin. The findings suggest that muscle fibers respond to the presence of cysts by expression of MHC I and II. The possible role of MHC I and II in the inflammatory response and on the cyst wall is also discussed.


Subject(s)
Inflammation/veterinary , Myositis/veterinary , Sarcocystis/classification , Sarcocystosis/veterinary , Sheep Diseases/pathology , Animals , Fluorescent Antibody Technique/veterinary , Immunohistochemistry/veterinary , Inflammation/parasitology , Inflammation/pathology , Major Histocompatibility Complex/immunology , Muscle, Skeletal/parasitology , Muscle, Skeletal/pathology , Myositis/parasitology , Myositis/pathology , Sarcocystis/genetics , Sarcocystis/isolation & purification , Sarcocystosis/parasitology , Sarcocystosis/pathology , Sheep , Sheep Diseases/parasitology , T-Lymphocytes/parasitology , T-Lymphocytes/pathology
7.
Proc Natl Acad Sci U S A ; 114(10): 2711-2716, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28223498

ABSTRACT

A live-attenuated malaria vaccine, Plasmodium falciparum sporozoite vaccine (PfSPZ Vaccine), confers sterile protection against controlled human malaria infection (CHMI) with Plasmodium falciparum (Pf) parasites homologous to the vaccine strain up to 14 mo after final vaccination. No injectable malaria vaccine has demonstrated long-term protection against CHMI using Pf parasites heterologous to the vaccine strain. Here, we conducted an open-label trial with PfSPZ Vaccine at a dose of 9.0 × 105 PfSPZ administered i.v. three times at 8-wk intervals to 15 malaria-naive adults. After CHMI with homologous Pf parasites 19 wk after final immunization, nine (64%) of 14 (95% CI, 35-87%) vaccinated volunteers remained without parasitemia compared with none of six nonvaccinated controls (P = 0.012). Of the nine nonparasitemic subjects, six underwent repeat CHMI with heterologous Pf7G8 parasites 33 wk after final immunization. Five (83%) of six (95% CI, 36-99%) remained without parasitemia compared with none of six nonvaccinated controls. PfSPZ-specific T-cell and antibody responses were detected in all vaccine recipients. Cytokine production by T cells from vaccinated subjects after in vitro stimulation with homologous (NF54) or heterologous (7G8) PfSPZ were highly correlated. Interestingly, PfSPZ-specific T-cell responses in the blood peaked after the first immunization and were not enhanced by subsequent immunizations. Collectively, these data suggest durable protection against homologous and heterologous Pf parasites can be achieved with PfSPZ Vaccine. Ongoing studies will determine whether protective efficacy can be enhanced by additional alterations in the vaccine dose and number of immunizations.


Subject(s)
Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Plasmodium falciparum/drug effects , Vaccines, Attenuated/administration & dosage , Adolescent , Adult , Female , Healthy Volunteers , Humans , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Male , Middle Aged , Plasmodium falciparum/pathogenicity , Sporozoites/immunology , Sporozoites/pathogenicity , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/parasitology , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/immunology
8.
Parasite Immunol ; 41(8): e12632, 2019 08.
Article in English | MEDLINE | ID: mdl-31099071

ABSTRACT

Trypanosoma brucei gambiense, an extracellular eukaryotic flagellate parasite, is the main etiological agent of human African trypanosomiasis (HAT) or sleeping sickness. Dendritic cells (DCs) play a pivotal role at the interface between innate and adaptive immune response and are implicated during HAT. In this study, we investigated the effects of T gambiense and its excreted/secreted factors (ESF) on the phenotype of human monocyte-derived DCs (Mo-DCs). Mo-DCs were cultured with trypanosomes, lipopolysaccharide (LPS), ESF derived from T gambiense bloodstream strain Biyamina (MHOM/SD/82), or both ESF and LPS. Importantly, ESF reduced the expression of the maturation markers HLA-DR and CD83, as well as the secretion of IL-12, TNF-alpha and IL-10, in LPS-stimulated Mo-DCs. During mixed-leucocyte reactions, LPS- plus ESF-exposed DCs induced a non-significant decrease in the IFN-gamma/IL-10 ratio of CD4 + T-cell cytokines. Based on the results presented here, we raise the hypothesis that T gambiense has developed an immune escape strategy through the secretion of paracrine mediators in order to limit maturation and activation of human DCs. The identification of the factor(s) in the T gambiense ESF and of the DCs signalling pathway(s) involved may be important in the development of new therapeutic targets.


Subject(s)
Dendritic Cells/immunology , Monocytes/immunology , Protozoan Proteins/immunology , Trypanosoma brucei gambiense/immunology , Trypanosomiasis, African/immunology , Animals , Dendritic Cells/parasitology , Female , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , Host-Parasite Interactions , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-12/genetics , Interleukin-12/immunology , Lipopolysaccharides/immunology , Mice , Monocytes/parasitology , Protozoan Proteins/genetics , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/parasitology , Trypanosoma brucei gambiense/genetics , Trypanosomiasis, African/genetics , Trypanosomiasis, African/parasitology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
9.
Infect Immun ; 86(1)2018 01.
Article in English | MEDLINE | ID: mdl-28993459

ABSTRACT

The histozoic myxozoan parasite Kudoa thyrsites causes postmortem myoliquefaction and is responsible for economic losses to salmon aquaculture in the Pacific Northwest. Despite its importance, little is known about the host-parasite relationship, including the host response to infection. The present work sought to characterize the immune response in Atlantic salmon during infection, recovery, and reexposure to K. thyrsites After exposure to infective seawater, infected and uninfected smolts were sampled three times over 4,275 degree-days. Histological analysis revealed infection severity decreased over time in exposed fish, while in controls there was no evidence of infection. Following a secondary exposure of all fish, severity of infection in the controls was similar to that measured in exposed fish at the first sampling time but was significantly reduced in reexposed fish, suggesting the acquisition of protective immunity. Using immunohistochemistry, we detected a population of MHIIß+ cells in infected muscle that followed a pattern of abundance concordant with parasite prevalence. Infiltration of these cells into infected myocytes preceded destruction of the plasmodium and dissemination of myxospores. Dual labeling indicated a majority of these cells were CD83+/MHIIß+ Using reverse transcription-quantitative PCR, we detected significant induction of cellular effectors, including macrophage/dendritic cells (mhii/cd83/mcsf), B cells (igm/igt), and cytotoxic T cells (cd8/nkl), in the musculature of infected fish. These data support a role for cellular effectors such as antigen-presenting cells (monocyte/macrophage and dendritic cells) along with B and T cells in the acquired protective immune response of Atlantic salmon against K. thyrsites.


Subject(s)
Adaptive Immunity/immunology , Antigen-Presenting Cells/immunology , Antigens, CD/immunology , Immunoglobulins/immunology , Membrane Glycoproteins/immunology , Myxozoa/immunology , Salmo salar/immunology , Salmo salar/parasitology , Salmon/immunology , Salmon/parasitology , Animals , Antigen-Presenting Cells/parasitology , Aquaculture/methods , B-Lymphocytes/immunology , B-Lymphocytes/parasitology , Dendritic Cells/immunology , Dendritic Cells/parasitology , Fish Diseases/immunology , Fish Diseases/parasitology , Host-Parasite Interactions/immunology , Macrophages/immunology , Macrophages/parasitology , Muscle Cells/immunology , Muscle Cells/parasitology , Muscle, Skeletal/immunology , Muscle, Skeletal/parasitology , Parasitic Diseases, Animal/immunology , Parasitic Diseases, Animal/parasitology , T-Lymphocytes/immunology , T-Lymphocytes/parasitology , CD83 Antigen
10.
Infect Immun ; 85(11)2017 11.
Article in English | MEDLINE | ID: mdl-28874445

ABSTRACT

Experimental cerebral malaria (ECM) is a gamma interferon (IFN-γ)-dependent syndrome. However, whether IFN-γ promotes ECM through direct and synergistic targeting of multiple cell populations or by acting primarily on a specific responsive cell type is currently unknown. Here, using a panel of cell- and compartment-specific IFN-γ receptor 2 (IFN-γR2)-deficient mice, we show that IFN-γ causes ECM by signaling within both the hematopoietic and nonhematopoietic compartments. Mechanistically, hematopoietic and nonhematopoietic compartment-specific IFN-γR signaling exerts additive effects in orchestrating intracerebral inflammation, leading to the development of ECM. Surprisingly, mice with specific deletion of IFN-γR2 expression on myeloid cells, T cells, or neurons were completely susceptible to terminal ECM. Utilizing a reductionist in vitro system, we show that synergistic IFN-γ and tumor necrosis factor (TNF) stimulation promotes strong activation of brain blood vessel endothelial cells. Combined, our data show that within the hematopoietic compartment, IFN-γ causes ECM by acting redundantly or by targeting non-T cell or non-myeloid cell populations. Within the nonhematopoietic compartment, brain endothelial cells, but not neurons, may be the major target of IFN-γ leading to ECM development. Collectively, our data provide information on how IFN-γ mediates the development of cerebral pathology during malaria infection.


Subject(s)
Brain/immunology , Endothelial Cells/immunology , Interferon-gamma/genetics , Malaria, Cerebral/genetics , Plasmodium berghei/pathogenicity , Receptors, Interferon/genetics , Animals , Brain/blood supply , Brain/parasitology , Brain/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Chemokines, CXC/genetics , Chemokines, CXC/immunology , Disease Models, Animal , Endothelial Cells/parasitology , Gene Expression Regulation , Interferon-gamma/immunology , Interleukins/genetics , Interleukins/immunology , Malaria, Cerebral/immunology , Malaria, Cerebral/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/parasitology , Neurons/immunology , Neurons/parasitology , Plasmodium berghei/immunology , Receptors, Interferon/deficiency , Receptors, Interferon/immunology , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/parasitology
11.
Gastric Cancer ; 20(3): 438-447, 2017 May.
Article in English | MEDLINE | ID: mdl-27318497

ABSTRACT

BACKGROUND: The neuronal splicing factor neuro-oncological ventral antigen 1 (NOVA1) is enriched in normal fibroblasts. Stromal spindle cells such as fibroblasts are major components of tissue inflammation and tertiary lymphoid structures within the microenvironment that contribute to the survival and growth of cancer cells. In the present study, we investigated changes of NOVA1 expression in tertiary lymphoid structures in early and advanced gastric cancer microenvironments in terms of tumor progression and immune regulation. METHODS: Using immunohistochemistry, we analyzed NOVA1 expression in tumor cells, T cells, and stromal spindle cells as well as infiltrating densities of CD3+ T cells, forkhead box P3 positive (FOXP3+) regulatory T cells, CD68+ macrophages, CD163+ M2 macrophages, and myeloperoxidase-positive neutrophils in 396 surgically resected gastric cancer tissues. RESULTS: Suppressed NOVA1 expression in tumor cells, T cells, and stromal spindle cells was closely related to decreased infiltration of FOXP3+ regulatory T cells, increased infiltration of CD68+ macrophages and CD163+ M2 macrophages, more advanced tumor stage, and inferior overall survival rate. In addition, low infiltration of CD3+ T cells and FOXP3+ regulatory T cells and high infiltration of CD68+ macrophages were associated with inferior overall survival. Specifically, weak NOVA1 expression in tumor cells was independently related to more advanced tumor stage and inferior overall survival. CONCLUSIONS: NOVA1 suppression was frequently noted in the gastric cancer microenvironment, and attenuated NOVA1 expression in tumor cells was associated with tumor progression and poor prognosis. This finding seems to be related to immune dysfunction through changes in the immune cell composition of T cells and macrophages.


Subject(s)
RNA-Binding Proteins/metabolism , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Tumor Microenvironment/immunology , Aged , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Female , Forkhead Transcription Factors/metabolism , Humans , Kaplan-Meier Estimate , Macrophages/metabolism , Macrophages/pathology , Male , Middle Aged , Neuro-Oncological Ventral Antigen , Stomach Neoplasms/mortality , Stromal Cells/metabolism , Stromal Cells/pathology , T-Lymphocytes/metabolism , T-Lymphocytes/parasitology , T-Lymphocytes, Regulatory/metabolism
12.
J Immunol ; 195(12): 5707-17, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26538396

ABSTRACT

Intracellular infections, such as those caused by the protozoan parasite Leishmania donovani, a causative agent of visceral leishmaniasis (VL), require a potent host proinflammatory response for control. IL-17 has emerged as an important proinflammatory cytokine required for limiting growth of both extracellular and intracellular pathogens. However, there are conflicting reports on the exact roles for IL-17 during parasitic infections and limited knowledge about cellular sources and the immune pathways it modulates. We examined the role of IL-17 in an experimental model of VL caused by infection of C57BL/6 mice with L. donovani and identified an early suppressive role for IL-17 in the liver that limited control of parasite growth. IL-17-producing γδ T cells recruited to the liver in the first week of infection were the critical source of IL-17 in this model, and CCR2(+) inflammatory monocytes were an important target for the suppressive effects of IL-17. Improved parasite control was independent of NO generation, but associated with maintenance of superoxide dismutase mRNA expression in the absence of IL-17 in the liver. Thus, we have identified a novel inhibitory function for IL-17 in parasitic infection, and our results demonstrate important interactions among γδ T cells, monocytes, and infected macrophages in the liver that can determine the outcome of parasitic infection.


Subject(s)
Interleukin-17/metabolism , Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , Liver/immunology , T-Lymphocytes/immunology , Animals , Disease Models, Animal , Humans , Immunosuppression Therapy , Leishmania donovani/growth & development , Liver/parasitology , Mice , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/parasitology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, CCR2/metabolism , Superoxide Dismutase/metabolism , T-Lymphocytes/parasitology
13.
J Biol Chem ; 290(32): 19496-511, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-25960542

ABSTRACT

The liver stages of the malaria parasite are clinically silent and constitute ideal targets for causal prophylactic drugs and vaccines. Cellular and molecular events responsible for liver stage development are poorly characterized. Here, we show that sporozoite, liver stage tryptophan-rich protein (SLTRiP) forms large multimers. Mice immunized with a purified recombinant SLTRiP protein gave high antibody titers in both inbred and outbred mice. Immunized mice showed highly significant levels of protection upon challenge with sporozoites and exhibited 10,000-fold fewer parasite 18S-rRNA copy numbers in their livers. The protection offered by immunization with SLTRiP came mainly from T-cells, and antibodies had little role to play despite their high titers. Immunofluorescence assays showed that SLTRiP is expressed in the sporozoite and early to late liver stages of malaria parasites. SLTRiP protein is exported to the cytosol of infected host cells during the early hours of parasite infection. Parasites deficient in SLTRiP were moderately defective in liver stage parasite development. A transcriptome profile of SLTRiP-deficient parasite-infected hepatocytes highlighted that SLTRiP interferes with multiple pathways in the host cell. We have demonstrated a role for SLTRiP in sporozoites and the liver stage of malaria parasites.


Subject(s)
Immunity, Cellular , Malaria Vaccines/immunology , Malaria/prevention & control , Plasmodium berghei/immunology , Protozoan Proteins/immunology , Sporozoites/immunology , T-Lymphocytes/drug effects , Amino Acid Sequence , Animals , Anopheles/parasitology , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Gene Expression , Immunization , Insect Vectors/parasitology , Liver/drug effects , Liver/immunology , Liver/parasitology , Liver/pathology , Malaria/immunology , Malaria/parasitology , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Plasmodium berghei/genetics , Protozoan Proteins/administration & dosage , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sequence Alignment , Sporozoites/chemistry , T-Lymphocytes/immunology , T-Lymphocytes/parasitology
14.
J Clin Immunol ; 36(1): 85-94, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26644312

ABSTRACT

PURPOSE: Leishmania, the causative organisms for leishmaniasis, reside in host macrophages and survive by modulating the microbicidal pathways via attenuation of the oxidative burst and/or suppression of cell-mediated immunity. As post kala-azar dermal leishmaniasis (PKDL), the dermal sequela of visceral leishmaniasis, has no animal model, the underlying mechanism(s) that nullify the microbicidal effector mechanisms remain poorly understood. This study was aimed at assessing the status of dipeptidyl peptidase CD26, a co-stimulatory molecule that is essential for T-cell signal activation. METHODS: The frequency/expression of CD26 and CD45RO/RA was evaluated by flow cytometry, while levels of soluble CD26 (sCD26), CXCL-10, RANTES, IL-10 and TGF-ß along with adenosine deaminase (ADA) activity were measured using ELISA. RESULTS: In patients with PKDL vis-à-vis healthy individuals, there was a significant decrease in the frequency and expression of CD26 on CD3(+)CD8(+) T-cells, which was accompanied by a significant lowering of plasma levels of sCD26. Furthermore, these patients showed a significant decrease in the frequency of CD45RO(+)/CD8(+) T-cells, concomitant with a significant increase in the proportion of CD45RA(+)/CD8(+) T-cells. This could collectively translate into reduced formation of the immunological synapse of CD26, CD45RO, and ADA, and lead to an attenuation of the Th1 responses. The decreased levels of CD26 and sCD26 correlated negatively with raised levels of Th2 cytokines, IL-10, and TGF-ß along with the lesional parasite load, indicating disease specificity. CONCLUSIONS: Taken together, the decreased expression and secretion of CD26 in patients with PKDL resulted in impairment of the CD26-ADA interaction, and thereby possibly contributed to T-cell unresponsiveness, emphasizing the need to develop immunomodulatory therapies against PKDL and by extension, the leishmaniases.


Subject(s)
Dipeptidyl Peptidase 4/metabolism , Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , Skin/immunology , T-Lymphocytes/immunology , Adolescent , Adult , Animals , Antigens, Protozoan/immunology , Cells, Cultured , Cytokines/metabolism , Dipeptidyl Peptidase 4/genetics , Female , Humans , India , Male , Skin/parasitology , T-Lymphocytes/parasitology , Th1-Th2 Balance , Young Adult
15.
Parasite Immunol ; 38(7): 414-8, 2016 07.
Article in English | MEDLINE | ID: mdl-27120498

ABSTRACT

Patients with cystic echinococcosis (CE) can harbour cysts for years or even decades, apparently without effect of the immune system on the metacestode. Although several immune evasion mechanisms by echinococcal cysts have been described, it is unclear whether the human leucocyte antigen (HLA) system plays a role in the susceptibility or resistance to CE in humans. HLA-G molecules are known to exert a suppressive action on dendritic cells maturation and on natural killer (NK) cells functions, therefore hampering T-cell responses and NK cytolysis. HLA-G plays an important role in immune tolerance, is involved in foetus and in allotransplant tolerance, and may be involved in tumoral and viral immune evasion. In this study, we assessed the presence and levels of soluble HLA-G (sHLA-G) in patients with CE using a commercial ELISA kit to determine whether host's HLA-G may have a role in the course of human CE.


Subject(s)
Echinococcosis/immunology , Echinococcus/growth & development , Echinococcus/immunology , HLA-G Antigens/immunology , Immune Evasion , Adult , Animals , Dendritic Cells/immunology , Dendritic Cells/parasitology , Echinococcosis/blood , Echinococcosis/parasitology , Echinococcus/genetics , Enzyme-Linked Immunosorbent Assay , Female , HLA-G Antigens/blood , Humans , Immune Tolerance , Killer Cells, Natural/immunology , Killer Cells, Natural/parasitology , Male , Middle Aged , T-Lymphocytes/immunology , T-Lymphocytes/parasitology , Young Adult
16.
Malar J ; 15: 201, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27068454

ABSTRACT

BACKGROUND: Plasmodium circumsporozoite protein (CSP) is a major surface antigen present in the sporozoite (Spz) stage of a malaria parasite. RTS, S vaccine, the most clinically advanced malaria vaccine, consists of a large portion of Plasmodium falciparum CSP (PfCSP). A highly infectious, recombinant rodent malaria, Plasmodium yoelii parasite bearing a full-length PfCSP, PfCSP/Py Spz, was needed as a tool to evaluate the role of PfCSP in mediating, protective, anti-malaria immunity in a mouse model. METHODS: A transgenic parasite, PfCSP/Py Spz, was generated by inserting a construct expressing the PfCSP at the locus of the P. yoelii CSP gene by double cross-over homologous recombination. Then the biological and protective properties of PfCSP/Py Spz were determined. RESULTS: This PfCSP/Py parasite produced up to 30,000 Spz in mosquito salivary glands, which is equal or even higher than the number of Spz produced by wild-type P. yoelii parasites. Five bites of PfCSP/Py-infected mosquitoes could induce blood infection in BALB/c mice. CONCLUSIONS: The current study has demonstrated a successful establishment of a transgenic P. yoelii parasite clone that is able to express a full-length PfCSP, PfCSP/Py parasite. Importantly, this PfCSP/Py parasite can be as infectious as the wild-type P. yoelii parasite both in mosquito vector and in mouse, a mammalian host. A new transgenic parasite that expresses a full-length PfCSP may become a useful tool for researchers to investigate immunity against PfCSP in a mouse model.


Subject(s)
Culicidae/parasitology , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Plasmodium yoelii/genetics , Plasmodium yoelii/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Animals , Antibodies, Protozoan/blood , Disease Models, Animal , Female , Mice , Mice, Inbred BALB C , Organisms, Genetically Modified/genetics , Organisms, Genetically Modified/immunology , Plasmodium falciparum/genetics , Salivary Glands/parasitology , T-Lymphocytes/parasitology , Vaccines, Synthetic/immunology
17.
Proc Natl Acad Sci U S A ; 110(15): 6055-60, 2013 Apr 09.
Article in English | MEDLINE | ID: mdl-23530242

ABSTRACT

Development of an antimalarial subunit vaccine inducing protective cytotoxic T lymphocyte (CTL)-mediated immunity could pave the way for malaria eradication. Experimental immunization with sporozoites induces this type of protective response, but the extremely large number of proteins expressed by Plasmodium parasites has so far prohibited the identification of sufficient discrete T-cell antigens to develop subunit vaccines that produce sterile immunity. Here, using mice singly immunized with Plasmodium yoelii sporozoites and high-throughput screening, we identified a unique CTL response against the parasite ribosomal L3 protein. Unlike CTL responses to the circumsporozoite protein (CSP), the population of L3-specific CTLs was not expanded by multiple sporozoite immunizations. CSP is abundant in the sporozoite itself, whereas L3 expression does not increase until the liver stage. The response induced by a single immunization with sporozoites reduces the parasite load in the liver so greatly during subsequent immunizations that L3-specific responses are only generated during the primary exposure. Functional L3-specific CTLs can, however, be expanded by heterologous prime-boost regimens. Thus, although repeat sporozoite immunization expands responses to preformed antigens like CSP that are present in the sporozoite itself, this immunization strategy may not expand CTLs targeting parasite proteins that are synthesized later. Heterologous strategies may be needed to increase CTL responses across the entire spectrum of Plasmodium liver-stage proteins.


Subject(s)
Plasmodium yoelii/immunology , Protozoan Proteins/immunology , Sporozoites/immunology , T-Lymphocytes/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Separation , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Immunization , Immunophenotyping , Interferon-gamma/metabolism , Malaria Vaccines/immunology , Mice , Mice, Inbred BALB C , Peptides/immunology , Protozoan Proteins/metabolism , Sequence Analysis, DNA , T-Lymphocytes/parasitology
18.
Trends Immunol ; 33(4): 181-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22398370

ABSTRACT

Helminth parasites survive through a combination of parasite longevity, repeated re-infection and selective immune suppression to prevent protective Th2 responses. To counteract helminth-induced immunosuppression, and to induce long-term immunological memory, understanding of the multiple regulatory pathways within the T cell compartment is needed. Extrinsic inhibition by regulatory T cells is a key element of Th2 suppression. In addition, Th2 cells in chronic regulatory environments become functionally impaired, indicating cell-intrinsic regulation, which compromises protective Th2 memory. We discuss these pathways and consider the potential for reversing unresponsiveness through stimulatory signals or replacement by new responder populations. Future vaccine or therapeutic strategies should aim to minimize extrinsic regulatory effects and simultaneously negate Th2 anergy to drive effector responses into a long-term functionally competent state.


Subject(s)
Helminthiasis/immunology , T-Lymphocytes/immunology , Adaptation, Biological , Animals , Cell Lineage , Forkhead Transcription Factors/immunology , Helminthiasis/parasitology , Humans , Immunologic Memory , T-Lymphocytes/cytology , T-Lymphocytes/parasitology
19.
Pharm Res ; 32(8): 2663-77, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25715698

ABSTRACT

PURPOSE: Since, Leishmania protozoans are obligate intracellular parasites of macrophages, an immunopotentiating macrophage-specific Amphotericin B (AB) delivery system would be ideally appropriate to increase its superiority for leishmaniasis treatment and to eliminate undesirable toxicity. Herein, we report AB entrapped mannose grafted chitosan nanocapsules (MnosCNc-AB) that results in effective treatment of visceral leishmaniasis, while also enhancing L. donovani specific T-cell immune responses in infected host. METHODS: MnosCNc-AB were prepared via synthesized mannosylated chitosan deposition on interface of oil/water nanoemulsion intermediate and were characterized. J774A.1 macrophage uptake potential, antileishmanial activity and immunomodulatory profile were evaluated in hamster. Tissue localization, biodistribution and toxicity profile were also investigated. RESULTS: MnosCNc-AB had nanometric size (197.8 ± 8.84 nm), unimodal distribution (0.115 ± 0.04), positive zeta potential (+31.7 ± 1.03 mV) and 97.5 ± 1.13% cargo encapsulation efficiency. Superior macrophage internalization of mannosylated chitosan nanocapsules compared to unmodified chitosan nanocapsules was observed by fluorescence-based assessment, further confirmed by rapid blood clearance and, greater localization and higher accumulation in macrophage rich liver and spleen. While, MnosCNc-AB mediated cargo distribution to kidney decreased. Augmented in vitro antileishmanial activity and in vivo pro-inflammatory mediator's expression were observed with MnosCNc-AB, led to significant reduction (∼90%) in splenic parasite burden. CONCLUSIONS: Results demonstrated that mannose ligand grafted chitosan nanocapsules could improve selective delivery of AB into macrophages via interactions with overexpressed mannose receptors thus reduce undesirable toxicity. Study provides evidence for MnosCNc-AB potential to leishmaniasis therapeutics and presents valuable therapeutic strategies for combating chronic macrophage-resident microbial infections.


Subject(s)
Antiprotozoal Agents/pharmacology , Lectins, C-Type/drug effects , Macrophages/metabolism , Macrophages/parasitology , Mannose-Binding Lectins/drug effects , Receptors, Cell Surface/drug effects , Amphotericin B/administration & dosage , Amphotericin B/pharmacology , Amphotericin B/therapeutic use , Animals , Antiprotozoal Agents/administration & dosage , Antiprotozoal Agents/pharmacokinetics , Body Burden , Chemistry, Pharmaceutical , Cricetinae , Drug Compounding , Drug Delivery Systems , Leishmania donovani/drug effects , Leishmaniasis, Visceral/drug therapy , Leishmaniasis, Visceral/psychology , Mannose Receptor , Mesocricetus , Mice , Nanocapsules , Particle Size , Rats , Rats, Wistar , Spleen/parasitology , T-Lymphocytes/drug effects , T-Lymphocytes/parasitology , Tissue Distribution
20.
J Immunol ; 191(1): 262-73, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23729437

ABSTRACT

A differential behavior among infected and bystander dendritic cells (DCs) has been explored in different infection models. We have analyzed both populations sorted on contact with visceral Leishmania infantum on a susceptible mice model evaluating the subsequent repercussions on adaptive immune response. Our results demonstrate a clear dichotomy between the immunomodulatory abilities of bystander and infected DCs. The bystander population presents increased levels of IL-12p40 and costimulatory molecules being capable to induce CD4(+) T cell activation with immune protective capabilities. In contrast, infected DCs, which express lower costimulatory molecules and higher levels of IL-10, promote the development of Leishmania Ag-specific, nonprotective T-bet(+)IFN-γ(+)IL-10(+) CD4(+) T cells with an effector phenotype. This specific polarization was found to be dependent on IL-12p70. Splenic infected DCs recovered from chronic infected animals are similarly capable to polarize ex vivo syngeneic naive CD4(+) T cells toward a T-bet(+)IFN-γ(+)IL-10(+) phenotype. Further analysis revealed that only MHC class II(high)-infected DCs were responsible for this polarization. The adoptive transfer of such polarized CD4(+) T cells facilitates visceral leishmaniasis in BALB/c mice in a clear contrast with their counterpart generated with bystander DCs that significantly potentiate protection. Further, we demonstrated that CD4(+) T cells primed by infected DCs in an IL-10 free system, thus deprived of T-bet(+)IFN-γ(+)IL-10(+) population, restore the immune response and reduce parasite load, supporting a deleterious role of IFN-γ(+)IL-10(+) T cells in the maintenance of infection. Overall, our results highlight novel subversion mechanisms by which nonprotective T-bet(+)IFN-γ(+)IL-10(+) T cells are associated with chronicity and prolonged parasite persistence.


Subject(s)
Cell Polarity/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class II/biosynthesis , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Leishmaniasis, Visceral/immunology , T-Box Domain Proteins/biosynthesis , T-Lymphocytes/immunology , Animals , Cells, Cultured , Chronic Disease , Coculture Techniques , Dendritic Cells/metabolism , Dendritic Cells/parasitology , Host-Pathogen Interactions/immunology , Immunophenotyping , Interleukin-10/deficiency , Leishmania infantum/immunology , Leishmania infantum/pathogenicity , Leishmaniasis, Visceral/parasitology , Leishmaniasis, Visceral/prevention & control , Mice , Mice, Inbred BALB C , T-Lymphocytes/metabolism , T-Lymphocytes/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL