Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
PLoS Pathog ; 18(2): e1010339, 2022 02.
Article in English | MEDLINE | ID: mdl-35157735

ABSTRACT

Adoptive T-cell immunotherapy has provided promising results in the treatment of viral complications in humans, particularly in the context of immunocompromised patients who have exhausted all other clinical options. The capacity to expand T cells from healthy immune individuals is providing a new approach to anti-viral immunotherapy, offering rapid off-the-shelf treatment with tailor-made human leukocyte antigen (HLA)-matched T cells. While most of this research has focused on the treatment of latent viral infections, emerging evidence that SARS-CoV-2-specific T cells play an important role in protection against COVID-19 suggests that the transfer of HLA-matched allogeneic off-the-shelf virus-specific T cells could provide a treatment option for patients with active COVID-19 or at risk of developing COVID-19. We initially screened 60 convalescent individuals and based on HLA typing and T-cell response profile, 12 individuals were selected for the development of a SARS-CoV-2-specific T-cell bank. We demonstrate that these T cells are specific for up to four SARS-CoV-2 antigens presented by a broad range of both HLA class I and class II alleles. These T cells show consistent functional and phenotypic properties, display cytotoxic potential against HLA-matched targets and can recognize HLA-matched cells infected with different SARS-CoV-2 variants. These observations demonstrate a robust approach for the production of SARS-CoV-2-specific T cells and provide the impetus for the development of a T-cell repository for clinical assessment.


Subject(s)
HLA Antigens/immunology , Immunotherapy, Adoptive , SARS-CoV-2/immunology , T-Lymphocytes/immunology , Adult , Epitopes, T-Lymphocyte , Female , HEK293 Cells , Humans , Immunophenotyping , Male , Middle Aged , Young Adult
2.
Allergy ; 75(9): 2279-2293, 2020 09.
Article in English | MEDLINE | ID: mdl-32277487

ABSTRACT

BACKGROUND: Respiratory syncytial virus (RSV) affects most infants early in life and is associated with increased asthma risk. The specific mechanism remains unknown. OBJECTIVE: To investigate the role of uric acid (UA) and IL-1ß in RSV immunopathology and asthma predisposition. METHODS: Tracheal aspirates from human infants with and without RSV were collected and analyzed for pro-IL-1ß mRNA and protein to establish a correlation in human disease. Neonatal mouse models of RSV were employed, wherein mice infected at 6-7 days of life were analyzed at 8 days postinfection, 5 weeks postinfection, or after a chronic cockroach allergen asthma model. A xanthine oxidase inhibitor or IL-1 receptor antagonist was administered during RSV infection. RESULTS: Human tracheal aspirates from RSV-infected infants showed elevated pro-IL-1ß mRNA and protein. Inhibition of UA or IL-1ß during neonatal murine RSV infection decreased mucus production, reduced cellular infiltrates to the lung (especially ILC2s), and decreased type 2 immune responses. Inhibition of either UA or IL-1ß during RSV infection led to chronic reductions in pulmonary immune cell composition and reduced type 2 immune responses and reduced similar responses after challenge with cockroach antigen. CONCLUSIONS: Inhibiting UA and IL-1ß during RSV infection ameliorates RSV immunopathology, reduces the consequences of allergen-induced asthma, and presents new therapeutic targets to reduce early-life viral-induced asthma development.


Subject(s)
Asthma , Respiratory Syncytial Virus Infections , Animals , Immunity, Innate , Lung , Lymphocytes , Mice , Mice, Inbred BALB C , Respiratory Syncytial Viruses , Uric Acid
3.
J Immunol ; 200(4): 1443-1456, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29321276

ABSTRACT

Differentiation of CD4+ Th cells is critical for immunity to malaria. Several innate immune signaling pathways have been implicated in the detection of blood-stage Plasmodium parasites, yet their influence over Th cell immunity remains unclear. In this study, we used Plasmodium-reactive TCR transgenic CD4+ T cells, termed PbTII cells, during nonlethal P. chabaudi chabaudi AS and P. yoelii 17XNL infection in mice, to examine Th cell development in vivo. We found no role for caspase1/11, stimulator of IFN genes, or mitochondrial antiviral-signaling protein, and only modest roles for MyD88 and TRIF-dependent signaling in controlling PbTII cell expansion. In contrast, IFN regulatory factor 3 (IRF3) was important for supporting PbTII expansion, promoting Th1 over T follicular helper (Tfh) differentiation, and controlling parasites during the first week of infection. IRF3 was not required for early priming by conventional dendritic cells, but was essential for promoting CXCL9 and MHC class II expression by inflammatory monocytes that supported PbTII responses in the spleen. Thereafter, IRF3-deficiency boosted Tfh responses, germinal center B cell and memory B cell development, parasite-specific Ab production, and resolution of infection. We also noted a B cell-intrinsic role for IRF3 in regulating humoral immune responses. Thus, we revealed roles for IRF3 in balancing Th1- and Tfh-dependent immunity during nonlethal infection with blood-stage Plasmodium parasites.


Subject(s)
Cell Differentiation/immunology , Interferon Regulatory Factor-3/immunology , Malaria/immunology , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/immunology , Animals , Female , Germinal Center/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Spleen/immunology
4.
Proc Natl Acad Sci U S A ; 114(29): 7701-7706, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28673996

ABSTRACT

Severe malaria and associated high parasite burdens occur more frequently in humans lacking robust adaptive immunity to Plasmodium falciparum Nevertheless, the host may partly control blood-stage parasite numbers while adaptive immunity is gradually established. Parasite control has typically been attributed to enhanced removal of parasites by the host, although in vivo quantification of this phenomenon remains challenging. We used a unique in vivo approach to determine the fate of a single cohort of semisynchronous, Plasmodium berghei ANKA- or Plasmodium yoelii 17XNL-parasitized red blood cells (pRBCs) after transfusion into naive or acutely infected mice. As previously shown, acutely infected mice, with ongoing splenic and systemic inflammatory responses, controlled parasite population growth more effectively than naive controls. Surprisingly, however, this was not associated with accelerated removal of pRBCs from circulation. Instead, transfused pRBCs remained in circulation longer in acutely infected mice. Flow cytometric assessment and mathematical modeling of intraerythrocytic parasite development revealed an unexpected and substantial slowing of parasite maturation in acutely infected mice, extending the life cycle from 24 h to 40 h. Importantly, impaired parasite maturation was the major contributor to control of parasite growth in acutely infected mice. Moreover, by performing the same experiments in rag1-/- mice, which lack T and B cells and mount weak inflammatory responses, we revealed that impaired parasite maturation is largely dependent upon the host response to infection. Thus, impairment of parasite maturation represents a host-mediated, immune system-dependent mechanism for limiting parasite population growth during the early stages of an acute blood-stage Plasmodium infection.


Subject(s)
Host-Parasite Interactions , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Plasmodium berghei/physiology , Plasmodium falciparum/physiology , Adaptive Immunity , Animals , Cytokines/metabolism , Erythrocytes/parasitology , Female , Flow Cytometry , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Immune System , Inflammation , Malaria , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Theoretical , Plasmodium yoelii/physiology
5.
PLoS Pathog ; 12(1): e1005398, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26765224

ABSTRACT

Tumor necrosis factor (TNF) is critical for controlling many intracellular infections, but can also contribute to inflammation. It can promote the destruction of important cell populations and trigger dramatic tissue remodeling following establishment of chronic disease. Therefore, a better understanding of TNF regulation is needed to allow pathogen control without causing or exacerbating disease. IL-10 is an important regulatory cytokine with broad activities, including the suppression of inflammation. IL-10 is produced by different immune cells; however, its regulation and function appears to be cell-specific and context-dependent. Recently, IL-10 produced by Th1 (Tr1) cells was shown to protect host tissues from inflammation induced following infection. Here, we identify a novel pathway of TNF regulation by IL-10 from Tr1 cells during parasitic infection. We report elevated Blimp-1 mRNA levels in CD4+ T cells from visceral leishmaniasis (VL) patients, and demonstrate IL-12 was essential for Blimp-1 expression and Tr1 cell development in experimental VL. Critically, we show Blimp-1-dependent IL-10 production by Tr1 cells prevents tissue damage caused by IFNγ-dependent TNF production. Therefore, we identify Blimp-1-dependent IL-10 produced by Tr1 cells as a key regulator of TNF-mediated pathology and identify Tr1 cells as potential therapeutic tools to control inflammation.


Subject(s)
Inflammation/immunology , Interleukin-10/biosynthesis , Leishmaniasis, Visceral/immunology , Repressor Proteins/immunology , Th1 Cells/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Disease Models, Animal , Female , Flow Cytometry , Humans , Inflammation/pathology , Interleukin-10/immunology , Leishmaniasis, Visceral/pathology , Malaria/immunology , Malaria/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Fluorescence , Positive Regulatory Domain I-Binding Factor 1 , T-Lymphocytes, Regulatory/immunology
6.
PLoS Pathog ; 12(11): e1005999, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27812214

ABSTRACT

Parasite-specific antibodies protect against blood-stage Plasmodium infection. However, in malaria-endemic regions, it takes many months for naturally-exposed individuals to develop robust humoral immunity. Explanations for this have focused on antigenic variation by Plasmodium, but have considered less whether host production of parasite-specific antibody is sub-optimal. In particular, it is unclear whether host immune factors might limit antibody responses. Here, we explored the effect of Type I Interferon signalling via IFNAR1 on CD4+ T-cell and B-cell responses in two non-lethal murine models of malaria, P. chabaudi chabaudi AS (PcAS) and P. yoelii 17XNL (Py17XNL) infection. Firstly, we demonstrated that CD4+ T-cells and ICOS-signalling were crucial for generating germinal centre (GC) B-cells, plasmablasts and parasite-specific antibodies, and likewise that T follicular helper (Tfh) cell responses relied on B cells. Next, we found that IFNAR1-signalling impeded the resolution of non-lethal blood-stage infection, which was associated with impaired production of parasite-specific IgM and several IgG sub-classes. Consistent with this, GC B-cell formation, Ig-class switching, plasmablast and Tfh differentiation were all impaired by IFNAR1-signalling. IFNAR1-signalling proceeded via conventional dendritic cells, and acted early by limiting activation, proliferation and ICOS expression by CD4+ T-cells, by restricting the localization of activated CD4+ T-cells adjacent to and within B-cell areas of the spleen, and by simultaneously suppressing Th1 and Tfh responses. Finally, IFNAR1-deficiency accelerated humoral immune responses and parasite control by boosting ICOS-signalling. Thus, we provide evidence of a host innate cytokine response that impedes the onset of humoral immunity during experimental malaria.


Subject(s)
Antibodies, Protozoan/immunology , Immunity, Humoral/immunology , Inducible T-Cell Co-Stimulator Protein/immunology , Malaria/immunology , Receptor, Interferon alpha-beta/immunology , Animals , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Plasmodium chabaudi/immunology , Plasmodium yoelii/immunology , Signal Transduction/immunology
7.
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
8.
Eur J Immunol ; 45(1): 130-41, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25319247

ABSTRACT

Type I IFN signaling suppresses splenic T helper 1 (Th1) responses during blood-stage Plasmodium berghei ANKA (PbA) infection in mice, and is crucial for mediating tissue accumulation of parasites and fatal cerebral symptoms via mechanisms that remain to be fully characterized. Interferon regulatory factor 7 (IRF7) is considered to be a master regulator of type I IFN responses. Here, we assessed IRF7 for its roles during lethal PbA infection and nonlethal Plasmodium chabaudi chabaudi AS (PcAS) infection as two distinct models of blood-stage malaria. We found that IRF7 was not essential for tissue accumulation of parasites, cerebral symptoms, or brain pathology. Using timed administration of anti-IFNAR1 mAb, we show that late IFNAR1 signaling promotes fatal disease via IRF7-independent mechanisms. Despite this, IRF7 significantly impaired early splenic Th1 responses and limited control of parasitemia during PbA infection.  Finally, IRF7 also suppressed antiparasitic immunity and Th1 responses during nonlethal PcAS infection. Together, our data support a model in which IRF7 suppresses antiparasitic immunity in the spleen, while IFNAR1-mediated, but IRF7-independent, signaling contributes to pathology in the brain during experimental blood-stage malaria.


Subject(s)
Brain/immunology , Interferon Regulatory Factor-7/immunology , Malaria, Cerebral/immunology , Receptor, Interferon alpha-beta/immunology , Spleen/immunology , Th1 Cells/immunology , Animals , Antibodies, Monoclonal/pharmacology , Brain/drug effects , Brain/parasitology , Disease Susceptibility , Erythrocytes/parasitology , Female , Gene Expression Regulation , Host-Parasite Interactions , Interferon Regulatory Factor-7/genetics , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Plasmodium berghei/immunology , Plasmodium chabaudi/immunology , Receptor, Interferon alpha-beta/antagonists & inhibitors , Receptor, Interferon alpha-beta/genetics , Signal Transduction , Spleen/drug effects , Spleen/parasitology , Th1 Cells/parasitology , Time Factors
9.
J Immunol ; 192(8): 3709-18, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24634490

ABSTRACT

Organ-specific immunity is a feature of many infectious diseases, including visceral leishmaniasis caused by Leishmania donovani. Experimental visceral leishmaniasis in genetically susceptible mice is characterized by an acute, resolving infection in the liver and chronic infection in the spleen. CD4+ T cell responses are critical for the establishment and maintenance of hepatic immunity in this disease model, but their role in chronically infected spleens remains unclear. In this study, we show that dendritic cells are critical for CD4+ T cell activation and expansion in all tissue sites examined. We found that FTY720-mediated blockade of T cell trafficking early in infection prevented Ag-specific CD4+ T cells from appearing in lymph nodes, but not the spleen and liver, suggesting that early CD4+ T cell priming does not occur in liver-draining lymph nodes. Extended treatment with FTY720 over the first month of infection increased parasite burdens, although this associated with blockade of lymphocyte egress from secondary lymphoid tissue, as well as with more generalized splenic lymphopenia. Importantly, we demonstrate that CD4+ T cells are required for the establishment and maintenance of antiparasitic immunity in the liver, as well as for immune surveillance and suppression of parasite outgrowth in chronically infected spleens. Finally, although early CD4+ T cell priming appeared to occur most effectively in the spleen, we unexpectedly revealed that protective CD4+ T cell-mediated hepatic immunity could be generated in the complete absence of all secondary lymphoid tissues.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunologic Memory , Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , Animals , Antigens, Protozoan/immunology , CD4-Positive T-Lymphocytes/drug effects , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/immunology , Female , Fingolimod Hydrochloride , Immunosuppressive Agents/pharmacology , Liver/drug effects , Liver/immunology , Liver/parasitology , Lymphocyte Activation/immunology , Lymphoid Tissue/drug effects , Lymphoid Tissue/immunology , Lymphoid Tissue/parasitology , Mice , Mice, Knockout , Propylene Glycols/pharmacology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Spleen/drug effects , Spleen/immunology , Spleen/parasitology
10.
Infect Immun ; 82(1): 212-20, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24144725

ABSTRACT

Parasite biomass and microvasculature obstruction are strongly associated with disease severity and death in Plasmodium falciparum-infected humans. This is related to sequestration of mature, blood-stage parasites (schizonts) in peripheral tissue. The prevailing view is that schizont sequestration leads to an increase in pathogen biomass, yet direct experimental data to support this are lacking. Here, we first studied parasite population dynamics in inbred wild-type (WT) mice infected with the rodent species of malaria, Plasmodium berghei ANKA. As is commonly reported, these mice became moribund due to large numbers of parasites in multiple tissues. We then studied infection dynamics in a genetically targeted line of mice, which displayed minimal tissue accumulation of parasites. We constructed a mathematical model of parasite biomass dynamics, incorporating schizont-specific host clearance, both with and without schizont sequestration. Combined use of mathematical and in vivo modeling indicated, first, that the slowing of parasite growth in the genetically targeted mice can be attributed to specific clearance of schizonts from the circulation and, second, that persistent parasite growth in WT mice can be explained solely as a result of schizont sequestration. Our work provides evidence that schizont sequestration could be a major biological process driving rapid, early increases in parasite biomass during blood-stage Plasmodium infection.


Subject(s)
Biomass , Erythrocytes/parasitology , Malaria/parasitology , Models, Biological , Plasmodium berghei/growth & development , Animals , Cytokines/metabolism , Disease Models, Animal , Erythrocytes/metabolism , Female , Flow Cytometry , Luminescent Measurements , Malaria/blood , Mice , Mice, Inbred C57BL , Plasmodium berghei/pathogenicity , Schizonts/parasitology
12.
J Immunol ; 186(11): 6148-56, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21525386

ABSTRACT

Parasite burden predicts disease severity in malaria and risk of death in cerebral malaria patients. In murine experimental cerebral malaria (ECM), parasite burden and CD8(+) T cells promote disease by mechanisms that are not fully understood. We found that the majority of brain-recruited CD8(+) T cells expressed granzyme B (GzmB). Furthermore, gzmB(-/-) mice harbored reduced parasite numbers in the brain as a consequence of enhanced antiparasitic CD4(+) T cell responses and were protected from ECM. We showed in these ECM-resistant mice that adoptively transferred, Ag-specific CD8(+) T cells migrated to the brain, but did not induce ECM until a critical Ag threshold was reached. ECM induction was exquisitely dependent on Ag-specific CD8(+) T cell-derived perforin and GzmB, but not IFN-γ. In wild-type mice, full activation of brain-recruited CD8(+) T cells also depended on a critical number of parasites in this tissue, which in turn, was sustained by these tissue-recruited cells. Thus, an interdependent relationship between parasite burden and CD8(+) T cells dictates the onset of perforin/GzmB-mediated ECM.


Subject(s)
Brain/immunology , CD8-Positive T-Lymphocytes/immunology , Granzymes/immunology , Malaria, Cerebral/immunology , Adoptive Transfer , Animals , Brain/metabolism , Brain/parasitology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cell Movement/immunology , Female , Flow Cytometry , Granzymes/genetics , Host-Parasite Interactions/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Malaria, Cerebral/parasitology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Perforin/immunology , Perforin/metabolism , Plasmodium chabaudi/immunology , Plasmodium chabaudi/physiology , Plasmodium yoelii/immunology , Plasmodium yoelii/physiology
13.
Eur J Immunol ; 41(9): 2688-98, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21674481

ABSTRACT

During blood-stage Plasmodium infection, large-scale invasion of RBCs often occurs before the generation of cellular immune responses. In Plasmodium berghei ANKA (PbA)-infected C57BL/6 mice, CD4(+) T cells controlled parasite numbers poorly, instead providing early help to pathogenic CD8(+) T cells. Expression analysis revealed that the transcriptional signature of CD4(+) T cells from PbA-infected mice was dominated by type I IFN (IFN-I) and IFN-γ-signalling pathway-related genes. A role for IFN-I during blood-stage Plasmodium infection had yet to be established. Here, we observed IFN-α protein production in the spleen of PbA-infected C57BL/6 mice over the first 2 days of infection. Mice deficient in IFN-I signalling had reduced parasite burdens, and displayed none of the fatal neurological symptoms associated with PbA infection. IFN-I substantially inhibited CD4(+) T-bet(+) T-cell-derived IFN-γ production, and prevented this emerging Th1 response from controlling parasites. Experiments using BM chimeric mice revealed that IFN-I signalled predominantly via radio-sensitive, haematopoietic cells, but did not suppress CD4(+) T cells via direct signalling to this cell type. Finally, we found that IFN-I suppressed IFN-γ production, and hampered efficient control of parasitaemia in mice infected with non-lethal Plasmodium chabaudi. Thus, we have elucidated a novel regulatory pathway in primary blood-stage Plasmodium infection that suppresses CD4(+) T-cell-mediated parasite control.


Subject(s)
Interferon Type I/metabolism , Malaria/immunology , Plasmodium berghei/immunology , Plasmodium chabaudi/immunology , Th1 Cells/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/parasitology , CD8-Positive T-Lymphocytes/pathology , Cells, Cultured , Immune Evasion , Immunosuppression Therapy , Interferon Type I/immunology , Interferon-gamma/metabolism , Life Cycle Stages , Mice , Mice, Inbred C57BL , Plasmodium berghei/pathogenicity , Plasmodium chabaudi/pathogenicity , Signal Transduction/immunology , T-Box Domain Proteins/metabolism , Th1 Cells/immunology , Th1 Cells/parasitology , Th1 Cells/pathology , Transplantation Chimera , Virulence
14.
PLoS Pathog ; 6(12): e1001221, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-21170302

ABSTRACT

Studies in malaria patients indicate that higher frequencies of peripheral blood CD4(+) Foxp3(+) CD25(+) regulatory T (Treg) cells correlate with increased blood parasitemia. This observation implies that Treg cells impair pathogen clearance and thus may be detrimental to the host during infection. In C57BL/6 mice infected with Plasmodium berghei ANKA, depletion of Foxp3(+) cells did not improve parasite control or disease outcome. In contrast, elevating frequencies of natural Treg cells in vivo using IL-2/anti-IL-2 complexes resulted in complete protection against severe disease. This protection was entirely dependent upon Foxp3(+) cells and resulted in lower parasite biomass, impaired antigen-specific CD4(+) T and CD8(+) T cell responses that would normally promote parasite tissue sequestration in this model, and reduced recruitment of conventional T cells to the brain. Furthermore, Foxp3(+) cell-mediated protection was dependent upon CTLA-4 but not IL-10. These data show that T cell-mediated parasite tissue sequestration can be reduced by regulatory T cells in a mouse model of malaria, thereby limiting malaria-induced immune pathology.


Subject(s)
Antigens, CD/pharmacology , Malaria, Cerebral/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/parasitology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/parasitology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/parasitology , CTLA-4 Antigen , Cell Proliferation , Forkhead Transcription Factors , Interleukin-10 , Malaria, Cerebral/prevention & control , Mice , Mice, Inbred C57BL , Plasmodium berghei
15.
Infect Immun ; 79(5): 1882-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21343349

ABSTRACT

Infection of C57BL/6 mice with Plasmodium berghei ANKA induces a fatal neurological disease commonly referred to as experimental cerebral malaria. The onset of neurological symptoms and mortality depend on pathogenic CD8(+) T cells and elevated parasite burdens in the brain. Here we provide clear evidence of liver damage in this model, which precedes and is independent of the onset of neurological symptoms. Large numbers of parasite-specific CD8(+) T cells accumulated in the liver following P. berghei ANKA infection. However, systemic depletion of these cells at various times during infection, while preventing neurological symptoms, failed to protect against liver damage or ameliorate it once established. In contrast, rapid, drug-mediated removal of parasites prevented hepatic injury if administered early and quickly resolved liver damage if administered after the onset of clinical symptoms. These data indicate that CD8(+) T cell-mediated immune pathology occurs in the brain but not the liver, while parasite-dependent pathology occurs in both organs during P. berghei ANKA infection. Therefore, we show that P. berghei ANKA infection of C57BL/6 mice is a multiorgan disease driven by the accumulation of parasites, which is also characterized by organ-specific CD8(+) T cell-mediated pathology.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Liver Diseases/parasitology , Malaria, Cerebral/pathology , Malaria, Cerebral/parasitology , Animals , Cell Separation , Female , Flow Cytometry , Liver Diseases/immunology , Liver Diseases/pathology , Malaria, Cerebral/immunology , Mice , Mice, Inbred C57BL , Plasmodium berghei/immunology
16.
J Exp Med ; 218(11)2021 11 01.
Article in English | MEDLINE | ID: mdl-34613328

ABSTRACT

Development of the immune system can be influenced by diverse extrinsic and intrinsic factors that influence the risk of disease. Severe early life respiratory syncytial virus (RSV) infection is associated with persistent immune alterations. Previously, our group had shown that adult mice orally supplemented with Lactobacillus johnsonii exhibited decreased airway immunopathology following RSV infection. Here, we demonstrate that offspring of mice supplemented with L. johnsonii exhibit reduced airway mucus and Th2 cell-mediated response to RSV infection. Maternal supplementation resulted in a consistent gut microbiome in mothers and their offspring. Importantly, supplemented maternal plasma and breastmilk, and offspring plasma, exhibited decreased inflammatory metabolites. Cross-fostering studies showed that prenatal Lactobacillus exposure led to decreased Th2 cytokines and lung inflammation following RSV infection, while postnatal Lactobacillus exposure diminished goblet cell hypertrophy and mucus production in the lung in response to airway infection. These studies demonstrate that Lactobacillus modulation of the maternal microbiome and associated metabolic reprogramming enhance airway protection against RSV in neonates.


Subject(s)
Gastrointestinal Microbiome/immunology , Respiratory Syncytial Virus Infections/immunology , Animals , Cytokines/immunology , Disease Models, Animal , Female , Lung/immunology , Lung/virology , Mice , Mice, Inbred BALB C , Pregnancy , Th2 Cells/immunology
17.
Mucosal Immunol ; 13(4): 691-701, 2020 07.
Article in English | MEDLINE | ID: mdl-32047272

ABSTRACT

Respiratory syncytial virus (RSV) infects a majority of infants and can cause severe disease leading to increased risk to develop asthma later in life. In the present studies we detected high levels of uric acid pathway components during RSV infection and examined whether they altered the pathogenesis of RSV infection. Inhibition of uric acid (UA) pathway activation during RSV infection in airway epithelial cells using XOI decreased the expression of IL-33, thymic stromal lymphopoietin (TSLP), and CCL2. In addition, treatment of RSV infected bone marrow-derived macrophages with XOI decreased production of IL-1ß. Thus, UA activation of different cell populations contributes different innate immune mediators that promote immunopathogenesis. When mice were treated with XOI or interleukin-1 receptor antagonist (IL1-ra) during RSV infection decreased pulmonary mucus was observed along with significantly reduced numbers of ILC2 and macrophages, accompanied by decreased IL-33 in bronchoalveolar lavage of the treated mice. These findings provide mechanistic insight into the development of RSV immunopathology and indicate that xanthine metabolites and UA are key immunoregulator molecules during RSV infection. Moreover, these findings suggest uric acid and IL-1ß as possible therapeutic targets to attenuate severe RSV disease.


Subject(s)
Cytokines/metabolism , Immunity, Innate , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Viruses/physiology , Th2 Cells/immunology , Th2 Cells/metabolism , Uric Acid/metabolism , Animals , Disease Models, Animal , Female , Humans , Inflammation Mediators/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Macrophages , Metabolic Networks and Pathways , Mice , Respiratory Mucosa/metabolism , Respiratory Syncytial Virus Infections/virology , Signal Transduction
18.
Cell Rep ; 30(8): 2512-2525.e9, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32101732

ABSTRACT

Type I interferons (IFNs) play critical roles in anti-viral and anti-tumor immunity. However, they also suppress protective immune responses in some infectious diseases. Here, we identify type I IFNs as major upstream regulators of CD4+ T cells from visceral leishmaniasis (VL) patients. Furthermore, we report that mice deficient in type I IFN signaling have significantly improved control of Leishmania donovani, a causative agent of human VL, associated with enhanced IFNγ but reduced IL-10 production by parasite-specific CD4+ T cells. Importantly, we identify a small-molecule inhibitor that can be used to block type I IFN signaling during established infection and acts synergistically with conventional anti-parasitic drugs to improve parasite clearance and enhance anti-parasitic CD4+ T cell responses in mice and humans. Thus, manipulation of type I IFN signaling is a promising strategy for improving disease outcome in VL patients.


Subject(s)
Immunity/drug effects , Interferon Type I/pharmacology , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/parasitology , Parasites/immunology , Amphotericin B/pharmacology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Epitopes , Humans , Inflammation/immunology , Inflammation/pathology , Interferon-gamma/pharmacology , Mice, Inbred C57BL , Nitriles , Parasites/drug effects , Pyrazoles/pharmacology , Pyrimidines , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/metabolism , Signal Transduction/drug effects
19.
Mucosal Immunol ; 12(2): 445-456, 2019 03.
Article in English | MEDLINE | ID: mdl-30617299

ABSTRACT

Stem cell factor (SCF) binds to the receptor c-Kit that is expressed on a number of myeloid and lymphoid cell populations, including Type 2 innate lymphoid cells (ILC2). However the importance of the SCF/c-Kit interaction in ILC2 has not been studied. Here we investigate the role of a specific SCF isoform, SCF248, in the allergic asthmatic response and SCF/c-Kit in ILC2 activation during chronic allergy. We observed that mice treated with a monoclonal antibody specific for SCF248 attenuated the development of chronic asthmatic disease by decreasing the number of mast cells, ILC2 and eosinophils, as well as reducing the accompanying pathogenic cytokine responses. These data were supported using SCFfl/fl-Col1-Cre-ERT mice and W/Wv mice that demonstrated the importance of the stem cell factor/c-Kit activation during chronic allergy and the accumulation of c-kit+ cells. Finally, these data demonstrate for the first time that SCF could activate ILC2 cells in vitro for the production of key allergic cytokines. Together these findings indicate that SCF is a critical cytokine involved in the activation of ILC2 that lead to more severe outcomes during chronic allergy and that the SCF248 isoform could be an important therapeutic target to control the disease progression.


Subject(s)
Asthma/immunology , Lung/pathology , Lymphocytes/immunology , Stem Cell Factor/metabolism , Allergens/immunology , Animals , Cells, Cultured , Chronic Disease , Collagen Type I/genetics , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Protein Isoforms/genetics , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/genetics , Stem Cell Factor/immunology , Th2 Cells/immunology
20.
Int J Parasitol ; 47(14): 913-922, 2017 12.
Article in English | MEDLINE | ID: mdl-28864033

ABSTRACT

The artemisinins are the first-line therapy for severe and uncomplicated malaria, since they cause rapid declines in parasitemia after treatment. Despite this, in vivo mechanisms underlying this rapid decline remain poorly characterised. The overall decline in parasitemia is the net effect of drug inhibition of parasites and host clearance, which competes against any ongoing parasite proliferation. Separating these mechanisms in vivo was not possible through measurements of total parasitemia alone. Therefore, we employed an adoptive transfer approach in which C57BL/6J mice were transfused with Plasmodium berghei ANKA strain-infected, fluorescent red blood cells, and subsequently drug-treated. This approach allowed us to distinguish between the initial drug-treated generation of parasites (Gen0), and their progeny (Gen1). Artesunate efficiently impaired maturation of Gen0 parasites, such that a sufficiently high dose completely arrested maturation after 6h of in vivo exposure. In addition, artesunate-affected parasites were cleared from circulation with a half-life of 6.7h. In vivo cell depletion studies using clodronate liposomes revealed an important role for host phagocytes in the removal of artesunate-affected parasites, particularly ring and trophozoite stages. Finally, we found that a second antimalarial drug, mefloquine, was less effective than artesunate at suppressing parasite maturation and driving host-mediated parasite clearance. Thus, we propose that in vivo artesunate treatment causes rapid decline in parasitemia by arresting parasite maturation and encouraging phagocyte-mediated clearance of parasitised RBCs.


Subject(s)
Antimalarials/pharmacology , Malaria/drug therapy , Parasitemia/drug therapy , Plasmodium berghei/drug effects , Plasmodium yoelii/drug effects , Adoptive Transfer , Animals , Antimalarials/administration & dosage , Artemisinins/administration & dosage , Artemisinins/pharmacology , Artesunate , Dose-Response Relationship, Drug , Erythrocytes/parasitology , Female , Flow Cytometry , Malaria/parasitology , Mefloquine/administration & dosage , Mefloquine/pharmacology , Mice , Mice, Inbred C57BL , Parasitemia/parasitology , Phagocytes , Plasmodium berghei/growth & development , Plasmodium yoelii/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL