Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(5): 114217, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38728141

ABSTRACT

While brain swelling, associated with fluid accumulation, is a known feature of pediatric cerebral malaria (CM), how fluid and macromolecules are drained from the brain during recovery from CM is unknown. Using the experimental CM (ECM) model, we show that fluid accumulation in the brain during CM is driven by vasogenic edema and not by perivascular cerebrospinal fluid (CSF) influx. We identify that fluid and molecules are removed from the brain extremely quickly in mice with ECM to the deep cervical lymph nodes (dcLNs), predominantly through basal routes and across the cribriform plate and the nasal lymphatics. In agreement, we demonstrate that ligation of the afferent lymphatic vessels draining to the dcLNs significantly impairs fluid drainage from the brain and lowers anti-malarial drug recovery from the ECM syndrome. Collectively, our results provide insight into the pathways that coordinate recovery from CM.


Subject(s)
Brain Edema , Malaria, Cerebral , Animals , Malaria, Cerebral/pathology , Mice , Disease Models, Animal , Lymphatic Vessels/metabolism , Mice, Inbred C57BL , Brain/pathology , Brain/parasitology , Brain/metabolism , Lymph Nodes/pathology , Plasmodium berghei , Female , Male
2.
Sci Adv ; 10(20): eadj3301, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38758780

ABSTRACT

Myeloid cells are highly prevalent in glioblastoma (GBM), existing in a spectrum of phenotypic and activation states. We now have limited knowledge of the tumor microenvironment (TME) determinants that influence the localization and the functions of the diverse myeloid cell populations in GBM. Here, we have utilized orthogonal imaging mass cytometry with single-cell and spatial transcriptomic approaches to identify and map the various myeloid populations in the human GBM tumor microenvironment (TME). Our results show that different myeloid populations have distinct and reproducible compartmentalization patterns in the GBM TME that is driven by tissue hypoxia, regional chemokine signaling, and varied homotypic and heterotypic cellular interactions. We subsequently identified specific tumor subregions in GBM, based on composition of identified myeloid cell populations, that were linked to patient survival. Our results provide insight into the spatial organization of myeloid cell subpopulations in GBM, and how this is predictive of clinical outcome.


Subject(s)
Glioblastoma , Myeloid Cells , Tumor Microenvironment , Glioblastoma/pathology , Glioblastoma/metabolism , Humans , Myeloid Cells/metabolism , Myeloid Cells/pathology , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/genetics , Cell Line, Tumor , Single-Cell Analysis , Hypoxia/metabolism , Gene Expression Profiling
3.
Brain Commun ; 5(4): fcad197, 2023.
Article in English | MEDLINE | ID: mdl-37680691

ABSTRACT

Bilateral vestibular schwannoma is the hallmark of NF2-related schwannomatosis, a rare tumour predisposition syndrome associated with a lifetime of surgical interventions, radiotherapy and off-label use of the anti-angiogenic drug bevacizumab. Unilateral vestibular schwannoma develops sporadically in non-NF2-related schwannomatosis patients for which there are no drug treatment options available. Tumour-infiltrating immune cells such as macrophages and T-cells correlate with increased vestibular schwannoma growth, which is suggested to be similar in sporadic and NF2-related schwannomatosis tumours. However, differences between NF2-related schwannomatosis and the more common sporadic disease include NF2-related schwannomatosis patients presenting an increased number of tumours, multiple tumour types and younger age at diagnosis. A comparison of the tumour microenvironment in sporadic and NF2-related schwannomatosis tumours is therefore required to underpin the development of immunotherapeutic targets, identify the possibility of extrapolating ex vivo data from sporadic vestibular schwannoma to NF2-related schwannomatosis and help inform clinical trial design with the feasibility of co-recruiting sporadic and NF2-related schwannomatosis patients. This study drew together bulk transcriptomic data from three published Affymetrix microarray datasets to compare the gene expression profiles of sporadic and NF2-related schwannomatosis vestibular schwannoma and subsequently deconvolved to predict the abundances of distinct tumour immune microenvironment populations. Data were validated using quantitative PCR and Hyperion imaging mass cytometry. Comparative bioinformatic analyses revealed close similarities in NF2-related schwannomatosis and sporadic vestibular schwannoma tumours across the three datasets. Significant inflammatory markers and signalling pathways were closely matched in NF2-related schwannomatosis and sporadic vestibular schwannoma, relating to the proliferation of macrophages, angiogenesis and inflammation. Bulk transcriptomic and imaging mass cytometry data identified macrophages as the most abundant immune population in vestibular schwannoma, comprising one-third of the cell mass in both NF2-related schwannomatosis and sporadic tumours. Importantly, there were no robust significant differences in signalling pathways, gene expression, cell type abundance or imaging mass cytometry staining between NF2-related schwannomatosis and sporadic vestibular schwannoma. These data indicate strong similarities in the tumour immune microenvironment of NF2-related schwannomatosis and sporadic vestibular schwannoma.

4.
PLoS One ; 18(5): e0285323, 2023.
Article in English | MEDLINE | ID: mdl-37141324

ABSTRACT

Monocytes contribute to the pro-inflammatory immune response during the blood stage of a Plasmodium falciparum infection, but their precise role in malaria pathology is not clear. Besides phagocytosis, monocytes are activated by products from P. falciparum infected erythrocytes (IE) and one of the activation pathways is potentially the NLR family pyrin domain containing 3 (NLRP3) inflammasome, a multi-protein complex that leads to the production of interleukin (IL)-1ß. In cerebral malaria cases, monocytes accumulate at IE sequestration sites in the brain microvascular and the locally produced IL-1ß, or other secreted molecules, could contribute to leakage of the blood-brain barrier. To study the activation of monocytes by IE within the brain microvasculature in an in vitro model, we co-cultured IT4var14 IE and the monocyte cell line THP-1 for 24 hours and determined whether generated soluble molecules affect barrier function of human brain microvascular endothelial cells, measured by real time trans-endothelial electrical resistance. The medium produced after co-culture did not affect endothelial barrier function and similarly no effect was measured after inducing oxidative stress by adding xanthine oxidase to the co-culture. While IL-1ß does decrease barrier function, barely any IL-1ß was produced in the co- cultures, indicative of a lack of or incomplete THP-1 activation by IE in this co-culture model.


Subject(s)
Malaria, Falciparum , Plasmodium falciparum , Humans , Monocytes/metabolism , Coculture Techniques , Endothelial Cells/metabolism , Inflammasomes/metabolism , Erythrocytes/metabolism , Cell Line , Brain/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Interleukin-1beta/metabolism
5.
Clin Immunol ; 249: 109287, 2023 04.
Article in English | MEDLINE | ID: mdl-36907540

ABSTRACT

Sensorineural hearing loss is the most common type of hearing loss in adults and occurs due to damage of the inner ear caused by a range of factors including ageing, excessive noise, toxins, and cancer. Auto-inflammatory disease is also a cause of hearing loss and there is evidence that inflammation could contribute to hearing loss in other conditions. Within the inner ear there are resident macrophage cells that respond to insults and whose activation correlates with damage. The NLRP3 inflammasome is a multi-molecular pro-inflammatory protein complex that forms in activated macrophages and may contribute to hearing loss. The aim of this article is to discuss the evidence for the NLRP3 inflammasome and associated cytokines as potential therapeutic targets for sensorineural hearing loss in conditions ranging from auto-inflammatory disease to tumour-induced hearing loss in vestibular schwannoma.


Subject(s)
Ear, Inner , Hearing Loss, Sensorineural , Hearing Loss , Adult , Humans , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Hearing Loss, Sensorineural/etiology , Hearing Loss, Sensorineural/metabolism , Ear, Inner/metabolism , Hearing Loss/complications
6.
Neurosurgery ; 92(3): 581-589, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36729787

ABSTRACT

BACKGROUND: There is evidence that macrophage infiltration in the tumor microenvironment promotes vestibular schwannoma (VS) growth. Efficacy of bevacizumab in NF2-associated VS demonstrates the value of therapies targeting the microvascular tumor microenvironment, and tumor-associated macrophages (TAMs) may represent another druggable target. OBJECTIVE: To characterize the relationship between growth, TAM infiltration, and circulating monocyte chemokines in a large cohort of patients with VS. METHODS: Immunostaining for Iba1 (macrophages), CD31 (endothelium), and fibrinogen (permeability) was performed on 101 growing and 19 static sporadic VS. The concentrations of monocyte-specific chemokines were measured in the plasma of 50 patients with growing VS and 25 patients with static VS. RESULTS: The Iba1 + cell count was significantly higher in growing as compared with static VS (592 vs 226/×20 HPF, P =<0.001). Similarly, the CD31 + % surface area was higher in growing VS (2.19% vs 1.32%, P = .01). There was a positive correlation between TAM infiltration and VS growth rate, which persisted after controlling for the effect of tumor volume (aR2 = 0.263, P =<0.001). The plasma concentrations of several monocytic chemokines were higher in patients with growing rather than static VS. CONCLUSION: There is a strong positive correlation between TAM infiltration and volumetric growth of VS, and this relationship is independent of tumor size. There is a colinear relationship between TAM infiltration and tumor vascularity, implying that inflammation and angiogenesis are interlinked in VS. Chemokines known to induce monocyte chemotaxis are found in higher concentrations in patients with growing VS, suggestive of a potential pathophysiological mechanism.


Subject(s)
Neuroma, Acoustic , Humans , Neuroma, Acoustic/pathology , Chemokines/metabolism , Inflammation/metabolism , Macrophages/metabolism , Macrophages/pathology , Tumor Microenvironment
7.
Malar J ; 21(1): 206, 2022 Jun 29.
Article in English | MEDLINE | ID: mdl-35768835

ABSTRACT

BACKGROUND: Rapid emergence of Plasmodium resistance to anti-malarial drug mainstays has driven a continual effort to discover novel drugs that target different biochemical pathway (s) during infection. Plasma membrane Calcium + 2 ATPase (PMCA4), a novel plasma membrane protein that regulates Calcium levels in various cells, namely red blood cell (RBC), endothelial cell and platelets, represents a new biochemical pathway that may interfere with susceptibility to malaria and/or severe malaria. METHODS: This study identified several pharmacological inhibitors of PMCA4, namely ATA and Resveratrol, and tested for their anti-malarial activities in vitro and in vivo using the Plasmodium falciparum 3D7 strain, the Plasmodium berghei ANKA strain, and Plasmodium yoelii 17XL strain as model. RESULTS: In vitro propagation of P. falciparum 3D7 strain in the presence of a wide concentration range of the inhibitors revealed that the parasite growth was inhibited in a dose-dependent manner, with IC50s at 634 and 0.231 µM, respectively. RESULTS: The results confirmed that both compounds exhibit moderate to potent anti-malarial activities with the strongest parasite growth inhibition shown by resveratrol at 0.231 µM. In vivo models using P. berghei ANKA for experimental cerebral malaria and P. yoelii 17XL for the effect on parasite growth, showed that the highest dose of ATA, 30 mg/kg BW, increased survival of the mice. Likewise, resveratrol inhibited the parasite growth following 4 days intraperitoneal injection at the dose of 100 mg/kg BW. CONCLUSION: The findings indicate that the PMCA4 of the human host may be a potential target for novel anti-malarials, either as single drug or in combination with the currently available effective anti-malarials.


Subject(s)
Antimalarials , Malaria, Cerebral , Parasites , Animals , Calcium/pharmacology , Mice , Plasma Membrane Calcium-Transporting ATPases , Plasmodium berghei , Plasmodium falciparum , Resveratrol/pharmacology
8.
Immunology ; 164(4): 737-753, 2021 12.
Article in English | MEDLINE | ID: mdl-34407221

ABSTRACT

Experimental cerebral malaria (ECM) is a severe complication of Plasmodium berghei ANKA (PbA) infection in mice, characterized by CD8+ T-cell accumulation within the brain. Whilst the dynamics of CD8+ T-cell activation and migration during extant primary PbA infection have been extensively researched, the fate of the parasite-specific CD8+ T cells upon resolution of ECM is not understood. In this study, we show that memory OT-I cells persist systemically within the spleen, lung and brain following recovery from ECM after primary PbA-OVA infection. Whereas memory OT-I cells within the spleen and lung exhibited canonical central memory (Tcm) and effector memory (Tem) phenotypes, respectively, memory OT-I cells within the brain post-PbA-OVA infection displayed an enriched CD69+ CD103- profile and expressed low levels of T-bet. OT-I cells within the brain were excluded from short-term intravascular antibody labelling but were targeted effectively by longer-term systemically administered antibodies. Thus, the memory OT-I cells were extravascular within the brain post-ECM but were potentially not resident memory cells. Importantly, whilst memory OT-I cells exhibited strong reactivation during secondary PbA-OVA infection, preventing activation of new primary effector T cells, they had dampened reactivation during a fourth PbA-OVA infection. Overall, our results demonstrate that memory CD8+ T cells are systemically distributed but exhibit a unique phenotype within the brain post-ECM, and that their reactivation characteristics are shaped by infection history. Our results raise important questions regarding the role of distinct memory CD8+ T-cell populations within the brain and other tissues during repeat Plasmodium infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Host-Parasite Interactions/immunology , Malaria/immunology , Malaria/parasitology , Plasmodium berghei/physiology , Animals , Biomarkers , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Chemotaxis, Leukocyte/immunology , Disease Susceptibility , Epitopes, T-Lymphocyte/immunology , Erythrocytes/immunology , Erythrocytes/parasitology , Extracellular Matrix , Immunologic Memory , Immunophenotyping , Life Cycle Stages , Lymphocyte Activation/immunology , Malaria/metabolism , Malaria/pathology , Malaria, Cerebral/immunology , Malaria, Cerebral/metabolism , Malaria, Cerebral/parasitology , Mice , Mice, Transgenic , Organ Specificity/immunology
9.
Malar J ; 20(1): 297, 2021 Jul 02.
Article in English | MEDLINE | ID: mdl-34215257

ABSTRACT

BACKGROUND: Recent genome wide analysis studies have identified a strong association between single nucleotide variations within the human ATP2B4 gene and susceptibility to severe malaria. The ATP2B4 gene encodes the plasma membrane calcium ATPase 4 (PMCA4), which is responsible for controlling the physiological level of intracellular calcium in many cell types, including red blood cells (RBCs). It is, therefore, postulated that genetic differences in the activity or expression level of PMCA4 alters intracellular Ca2+ levels and affects RBC hydration, modulating the invasion and growth of the Plasmodium parasite within its target host cell. METHODS: In this study the course of three different Plasmodium spp. infections were examined in mice with systemic knockout of Pmca4 expression. RESULTS: Ablation of PMCA4 reduced the size of RBCs and their haemoglobin content but did not affect RBC maturation and reticulocyte count. Surprisingly, knockout of PMCA4 did not significantly alter peripheral parasite burdens or the dynamics of blood stage Plasmodium chabaudi infection or reticulocyte-restricted Plasmodium yoelii infection. Interestingly, although ablation of PMCA4 did not affect peripheral parasite levels during Plasmodium berghei infection, it did promote slight protection against experimental cerebral malaria, associated with a minor reduction in antigen-experienced T cell accumulation in the brain. CONCLUSIONS: The finding suggests that PMCA4 may play a minor role in the development of severe malarial complications, but that this appears independent of direct effects on parasite invasion, growth or survival within RBCs.


Subject(s)
Disease Resistance/genetics , Malaria/genetics , Plasma Membrane Calcium-Transporting ATPases/genetics , Plasmodium/physiology , Animals , Cell Membrane , Malaria/blood , Malaria/parasitology , Malaria, Cerebral/genetics , Malaria, Cerebral/parasitology , Mice , Mice, Knockout , Plasma Membrane Calcium-Transporting ATPases/metabolism , Plasmodium berghei/physiology , Plasmodium chabaudi/physiology , Plasmodium yoelii/physiology
10.
J Immunol ; 205(6): 1608-1619, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32817333

ABSTRACT

CD4+ T cell functional inhibition (exhaustion) is a hallmark of malaria and correlates with impaired parasite control and infection chronicity. However, the mechanisms of CD4+ T cell exhaustion are still poorly understood. In this study, we show that Ag-experienced (Ag-exp) CD4+ T cell exhaustion during Plasmodium yoelii nonlethal infection occurs alongside the reduction in mammalian target of rapamycin (mTOR) activity and restriction in CD4+ T cell glycolytic capacity. We demonstrate that the loss of glycolytic metabolism and mTOR activity within the exhausted Ag-expCD4+ T cell population during infection coincides with reduction in T-bet expression. T-bet was found to directly bind to and control the transcription of various mTOR and metabolism-related genes within effector CD4+ T cells. Consistent with this, Ag-expTh1 cells exhibited significantly higher and sustained mTOR activity than effector T-bet- (non-Th1) Ag-expT cells throughout the course of malaria. We identified mTOR to be redundant for sustaining T-bet expression in activated Th1 cells, whereas mTOR was necessary but not sufficient for maintaining IFN-γ production by Th1 cells. Immunotherapy targeting PD-1, CTLA-4, and IL-27 blocked CD4+ T cell exhaustion during malaria infection and was associated with elevated T-bet expression and a concomitant increased CD4+ T cell glycolytic metabolism. Collectively, our data suggest that mTOR activity is linked to T-bet in Ag-expCD4+ T cells but that reduction in mTOR activity may not directly underpin Ag-expTh1 cell loss and exhaustion during malaria infection. These data have implications for therapeutic reactivation of exhausted CD4+ T cells during malaria infection and other chronic conditions.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immune Checkpoint Inhibitors/therapeutic use , Malaria/immunology , Mechanistic Target of Rapamycin Complex 1/metabolism , Plasmodium yoelii/physiology , T-Box Domain Proteins/metabolism , Th1 Cells/immunology , Animals , Cellular Senescence , Gene Expression Regulation , Glycolysis , Humans , Immune Tolerance , Immunologic Memory , Interferon-gamma/metabolism , Interleukin-27/metabolism , Lymphocyte Activation , Malaria/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Box Domain Proteins/genetics
11.
Parasite Immunol ; 42(9): e12723, 2020 09.
Article in English | MEDLINE | ID: mdl-32306409

ABSTRACT

AIMS: Co-inhibitory receptors play a major role in controlling the Th1 response during blood-stage malaria. Whilst PD-1 is viewed as the dominant co-inhibitory receptor restricting T cell responses, the roles of other such receptors in coordinating Th1 cell activity during malaria are poorly understood. METHODS AND RESULTS: Here, we show that the co-inhibitory receptor Tim-3 is expressed on splenic antigen-specific T-bet+ (Th1) OT-II cells transiently during the early stage of infection with transgenic Plasmodium yoelii NL parasites expressing ovalbumin (P yoelii NL-OVA). We reveal that co-blockade of Tim-3 and PD-L1 during the acute phase of P yoelii NL infection did not improve the Th1 cell response but instead led to a specific reduction in the numbers of splenic Th1 OT-II cells. Combined blockade of Tim-3 and PD-L1 did elevate anti-parasite IgG antibody responses. Nevertheless, co-blockade of Tim-3 and PD-L1 did not affect IFN-γ production by OT-II cells and did not influence parasite control during P yoelii NL-OVA infection. CONCLUSION: Thus, our results show that Tim-3 plays an unexpected combinatorial role with PD-1 in promoting and/ or sustaining a Th1 cell response during the early phase of blood-stage P. yoelii NL infection but combined blockade does not dramatically influence anti-parasite immunity.


Subject(s)
Hepatitis A Virus Cellular Receptor 2/immunology , Malaria/immunology , Programmed Cell Death 1 Receptor/immunology , Th1 Cells/immunology , Animals , B7-H1 Antigen , Cell Line , Epitopes/immunology , Malaria/parasitology , Male , Mice, Inbred C57BL , Spleen/immunology
12.
Front Immunol ; 10: 1747, 2019.
Article in English | MEDLINE | ID: mdl-31396236

ABSTRACT

CD8+ T cells have been shown to play a critical role in the pathogenesis of experimental cerebral malaria (ECM) in mice, but their role in development of human cerebral malaria (HCM) remains unclear. Thus, in this study we have provided the first direct contrast of the accumulation of CD8+ T cells in the brain during HCM and ECM. HCM cases were from children who died of Plasmodium falciparum cerebral malaria at Queen Elizabeth Central Hospital (Malawi) between 2003 and 2010. ECM was induced by infecting C57BL/6J mice with P. berghei ANKA. We demonstrate similarities in the intracerebral CD8+ T cell responses in ECM and HCM, in particular an apparent shared choroid plexus-meningeal route of CD8+ T cell accumulation in the brain. Nevertheless, we also reveal some potentially important differences in compartmentalization of CD8+ T cells within the cerebrovascular bed in HCM and ECM.


Subject(s)
Brain , CD8-Positive T-Lymphocytes , Malaria, Cerebral , Malaria, Falciparum , Plasmodium berghei/immunology , Plasmodium falciparum/immunology , Animals , Brain/immunology , Brain/parasitology , Brain/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Child , Child, Preschool , Female , Humans , Malaria, Cerebral/immunology , Malaria, Cerebral/pathology , Malaria, Falciparum/immunology , Malaria, Falciparum/pathology , Male , Mice
13.
Front Immunol ; 10: 248, 2019.
Article in English | MEDLINE | ID: mdl-30846985

ABSTRACT

Cerebral malaria (CM) is one of the most severe complications of Plasmodium falciparum infection. There is evidence that repeated parasite exposure promotes resistance against CM. However, the immunological basis of this infection-induced resistance remains poorly understood. Here, utilizing the Plasmodium berghei ANKA (PbA) model of experimental cerebral malaria (ECM), we show that three rounds of infection and drug-cure protects against the development of ECM during a subsequent fourth (4X) infection. Exposure-induced resistance was associated with specific suppression of CD8+ T cell activation and CTL-related pathways, which corresponded with the development of heterogeneous atypical B cell populations as well as the gradual infection-induced generation and maintenance of high levels of anti-parasite IgG. Mechanistically, transfer of high-titer anti-parasite IgG did not protect 1X infected mice against ECM and depletion of atypical and regulatory B cells during 4X infection failed to abrogate infection-induced resistance to ECM. However, IgMi mice that were unable to produce secreted antibody, or undergo class switching, during the repeated rounds of infection failed to develop resistance against ECM. The failure of infection-induced protection in IgMi mice was associated with impaired development of atypical B cell populations and the inability to suppress pathogenic CD8+ T cell responses. Our results, therefore, suggest the importance of anti-parasite antibody responses, gradually acquired, and maintained through repeated Plasmodium infections, for modulating the B cell compartment and eventually suppressing memory CD8+ T cell reactivation to establish infection-induced resistance to ECM.


Subject(s)
Antibody Formation/immunology , Brain/immunology , CD8-Positive T-Lymphocytes/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Animals , Brain/parasitology , CD8-Positive T-Lymphocytes/parasitology , Lymphocyte Activation/immunology , Malaria, Cerebral/parasitology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Mice , Mice, Inbred C57BL
14.
Front Immunol ; 10: 2842, 2019.
Article in English | MEDLINE | ID: mdl-31921120

ABSTRACT

How B cells contribute to protective immunity against parasitic nematodes remains unclear, with their importance as accessory cells underexplored. In this study, anti-CD20 monoclonal antibody (α-CD20 mAb)-mediated depletion of B cells from C57BL/6 mice revealed an important role for B cells in supporting Th2 immune responses and thus expulsion of Trichuris muris (T. muris). C57BL/6 mice normally mount mixed Th1/Th2 immune responses to T. muris and expel the parasite by the third week post infection. However, B cell-depleted C57BL/6 had significantly reduced Th2-type cytokines post infection and failed to expel the parasite. IFN-γ production in the MLN of C57BL/6 mice receiving α-CD20 mAb treatment was not affected, collectively resulting in an overall change in Th1/Th2 balance in favor of Th1. Further, the expression of IFN-γ and IFN-γ-induced genes at the effector site, the gut, was significantly increased in the absence of B cells. Interestingly, and in complete contrast, BALB/c mice, which mount strongly polarized Th2 immune responses, rather than mixed Th1/Th2 immune responses, were still able to expel T. muris in the absence of B cells. We thus hypothesized that the B cell plays a critical role in enabling strong Th2 responses in the context of mixed Th1/Th2 settings, with the role becoming redundant in highly Th2 polarized environments. In support of this, neutralization of IFN-γ in B cell depleted C57BL/6 restored resistance against T. muris infection. Thus, our data suggest an important role of B cells in supporting Th2-type immune responses in mixed IFN-γ-rich Th1/Th2 settings.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Host-Parasite Interactions/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Trichuriasis/immunology , Trichuris/immunology , Animals , Biomarkers , Cytokines/biosynthesis , Flow Cytometry , Gene Expression Regulation , Genetic Background , Host-Parasite Interactions/genetics , Immunohistochemistry , Male , Mice , Single-Cell Analysis , Trichuriasis/parasitology
15.
Proc Natl Acad Sci U S A ; 115(28): 7404-7409, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29954866

ABSTRACT

Cerebral malaria (CM) is a serious neurological complication caused by Plasmodium falciparum infection. Currently, the only treatment for CM is the provision of antimalarial drugs; however, such treatment by itself often fails to prevent death or development of neurological sequelae. To identify potential improved treatments for CM, we performed a nonbiased whole-brain transcriptomic time-course analysis of antimalarial drug chemotherapy of murine experimental CM (ECM). Bioinformatics analyses revealed IL33 as a critical regulator of neuroinflammation and cerebral pathology that is down-regulated in the brain during fatal ECM and in the acute period following treatment of ECM. Consistent with this, administration of IL33 alongside antimalarial drugs significantly improved the treatment success of established ECM. Mechanistically, IL33 treatment reduced inflammasome activation and IL1ß production in microglia and intracerebral monocytes in the acute recovery period following treatment of ECM. Moreover, treatment with the NLRP3-inflammasome inhibitor MCC950 alongside antimalarial drugs phenocopied the protective effect of IL33 therapy in improving the recovery from established ECM. We further showed that IL1ß release from macrophages was stimulated by hemozoin and antimalarial drugs and that this was inhibited by MCC950. Our results therefore demonstrate that manipulation of the IL33-NLRP3 axis may be an effective therapy to suppress neuroinflammation and improve the efficacy of antimalarial drug treatment of CM.


Subject(s)
Antimalarials/pharmacology , Brain/parasitology , Drug Delivery Systems/methods , Interleukin-33/metabolism , Malaria, Cerebral/drug therapy , Malaria, Falciparum/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Plasmodium falciparum/metabolism , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Female , Gene Expression Profiling , Hemeproteins/metabolism , Interleukin-1beta/biosynthesis , Interleukin-33/antagonists & inhibitors , Macrophages/metabolism , Macrophages/pathology , Malaria, Cerebral/metabolism , Malaria, Cerebral/pathology , Malaria, Falciparum/metabolism , Malaria, Falciparum/pathology , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Transcriptome/drug effects
16.
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
17.
PLoS Pathog ; 13(3): e1006267, 2017 03.
Article in English | MEDLINE | ID: mdl-28273147

ABSTRACT

The murine model of experimental cerebral malaria (ECM) has been utilised extensively in recent years to study the pathogenesis of human cerebral malaria (HCM). However, it has been proposed that the aetiologies of ECM and HCM are distinct, and, consequently, no useful mechanistic insights into the pathogenesis of HCM can be obtained from studying the ECM model. Therefore, in order to determine the similarities and differences in the pathology of ECM and HCM, we have performed the first spatial and quantitative histopathological assessment of the ECM syndrome. We demonstrate that the accumulation of parasitised red blood cells (pRBCs) in brain capillaries is a specific feature of ECM that is not observed during mild murine malaria infections. Critically, we show that individual pRBCs appear to occlude murine brain capillaries during ECM. As pRBC-mediated congestion of brain microvessels is a hallmark of HCM, this suggests that the impact of parasite accumulation on cerebral blood flow may ultimately be similar in mice and humans during ECM and HCM, respectively. Additionally, we demonstrate that cerebrovascular CD8+ T-cells appear to co-localise with accumulated pRBCs, an event that corresponds with development of widespread vascular leakage. As in HCM, we show that vascular leakage is not dependent on extensive vascular destruction. Instead, we show that vascular leakage is associated with alterations in transcellular and paracellular transport mechanisms. Finally, as in HCM, we observed axonal injury and demyelination in ECM adjacent to diverse vasculopathies. Collectively, our data therefore shows that, despite very different presentation, and apparently distinct mechanisms, of parasite accumulation, there appear to be a number of comparable features of cerebral pathology in mice and in humans during ECM and HCM, respectively. Thus, when used appropriately, the ECM model may be useful for studying specific pathological features of HCM.


Subject(s)
Brain/pathology , Brain/parasitology , Disease Models, Animal , Malaria, Cerebral/pathology , Malaria, Cerebral/parasitology , Animals , Erythrocytes/parasitology , Female , Fluorescent Antibody Technique , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Plasmodium berghei
18.
J Immunol ; 197(8): 3152-3164, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27630165

ABSTRACT

CD4+ T cells that produce IFN-γ are the source of host-protective IL-10 during primary infection with a number of different pathogens, including Plasmodium spp. The fate of these CD4+IFN-γ+IL-10+ T cells following clearance of primary infection and their subsequent influence on the course of repeated infections is, however, presently unknown. In this study, utilizing IFN-γ-yellow fluorescent protein (YFP) and IL-10-GFP dual reporter mice, we show that primary malaria infection-induced CD4+YFP+GFP+ T cells have limited memory potential, do not stably express IL-10, and are disproportionately lost from the Ag-experienced CD4+ T cell memory population during the maintenance phase postinfection. CD4+YFP+GFP+ T cells generally exhibited a short-lived effector rather than effector memory T cell phenotype postinfection and expressed high levels of PD-1, Lag-3, and TIGIT, indicative of cellular exhaustion. Consistently, the surviving CD4+YFP+GFP+ T cell-derived cells were unresponsive and failed to proliferate during the early phase of secondary infection. In contrast, CD4+YFP+GFP- T cell-derived cells expanded rapidly and upregulated IL-10 expression during secondary infection. Correspondingly, CD4+ T cells were the major producers within an accelerated and amplified IL-10 response during the early stage of secondary malaria infection. Notably, IL-10 exerted quantitatively stronger regulatory effects on innate and CD4+ T cell responses during primary and secondary infections, respectively. The results in this study significantly improve our understanding of the durability of IL-10-producing CD4+ T cells postinfection and provide information on how IL-10 may contribute to optimized parasite control and prevention of immune-mediated pathology during repeated malaria infections.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunologic Memory , Interleukin-10/metabolism , Malaria/immunology , Plasmodium/immunology , Animals , CD4-Positive T-Lymphocytes/parasitology , CD4-Positive T-Lymphocytes/transplantation , Cell Survival , Cells, Cultured , Humans , Immunosenescence , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-10/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic
19.
Infect Immun ; 84(1): 34-46, 2016 01.
Article in English | MEDLINE | ID: mdl-26459508

ABSTRACT

Immune-mediated pathology in interleukin-10 (IL-10)-deficient mice during blood-stage malaria infection typically manifests in nonlymphoid organs, such as the liver and lung. Thus, it is critical to define the cellular sources of IL-10 in these sensitive nonlymphoid compartments during infection. Moreover, it is important to determine if IL-10 production is controlled through conserved or disparate molecular programs in distinct anatomical locations during malaria infection, as this may enable spatiotemporal tuning of the regulatory immune response. In this study, using dual gamma interferon (IFN-γ)-yellow fluorescent protein (YFP) and IL-10-green fluorescent protein (GFP) reporter mice, we show that CD4(+) YFP(+) T cells are the major source of IL-10 in both lymphoid and nonlymphoid compartments throughout the course of blood-stage Plasmodium yoelii infection. Mature splenic CD4(+) YFP(+) GFP(+) T cells, which preferentially expressed high levels of CCR5, were capable of migrating to and seeding the nonlymphoid tissues, indicating that the systemically distributed host-protective cells have a common developmental history. Despite exhibiting comparable phenotypes, CD4(+) YFP(+) GFP(+) T cells from the liver and lung produced significantly larger quantities of IL-10 than their splenic counterparts, showing that the CD4(+) YFP(+) GFP(+) T cells exert graded functions in distinct tissue locations during infection. Unexpectedly, given the unique environmental conditions within discrete nonlymphoid and lymphoid organs, we show that IL-10 production by CD4(+) YFP(+) T cells is controlled systemically during malaria infection through IL-27 receptor signaling that is supported after CD4(+) T cell priming by ICOS signaling. The results in this study substantially improve our understanding of the systemic IL-10 response to malaria infection, particularly within sensitive nonlymphoid organs.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Inducible T-Cell Co-Stimulator Protein/immunology , Interleukin-10/immunology , Interleukins/immunology , Malaria/immunology , Adoptive Transfer , Animals , Bacterial Proteins/genetics , CD4-Positive T-Lymphocytes/transplantation , Green Fluorescent Proteins/genetics , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-10/genetics , Liver/immunology , Luminescent Proteins/genetics , Lung/immunology , Malaria/parasitology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plasmodium yoelii/immunology , Spleen/immunology
20.
PLoS Pathog ; 11(11): e1005210, 2015.
Article in English | MEDLINE | ID: mdl-26562533

ABSTRACT

There is significant evidence that brain-infiltrating CD8+ T cells play a central role in the development of experimental cerebral malaria (ECM) during Plasmodium berghei ANKA infection of C57BL/6 mice. However, the mechanisms through which they mediate their pathogenic activity during malaria infection remain poorly understood. Utilizing intravital two-photon microscopy combined with detailed ex vivo flow cytometric analysis, we show that brain-infiltrating T cells accumulate within the perivascular spaces of brains of mice infected with both ECM-inducing (P. berghei ANKA) and non-inducing (P. berghei NK65) infections. However, perivascular T cells displayed an arrested behavior specifically during P. berghei ANKA infection, despite the brain-accumulating CD8+ T cells exhibiting comparable activation phenotypes during both infections. We observed T cells forming long-term cognate interactions with CX3CR1-bearing antigen presenting cells within the brains during P. berghei ANKA infection, but abrogation of this interaction by targeted depletion of the APC cells failed to prevent ECM development. Pathogenic CD8+ T cells were found to colocalize with rare apoptotic cells expressing CD31, a marker of endothelial cells, within the brain during ECM. However, cellular apoptosis was a rare event and did not result in loss of cerebral vasculature or correspond with the extensive disruption to its integrity observed during ECM. In summary, our data show that the arrest of T cells in the perivascular compartments of the brain is a unique signature of ECM-inducing malaria infection and implies an important role for this event in the development of the ECM-syndrome.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Malaria, Cerebral/immunology , Malaria, Falciparum/microbiology , Parasitemia/immunology , Plasmodium berghei/immunology , Animals , CD8-Positive T-Lymphocytes/parasitology , Disease Models, Animal , Malaria, Cerebral/parasitology , Malaria, Cerebral/pathology , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...