Article

Perforin-Dependent Brain-Infiltrating Cytotoxic CD8+ T Lymphocytes Mediate Experimental Cerebral Malaria Pathogenesis

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract

Experimental cerebral malaria (ECM) resulting from Plasmodium berghei ANKA infection involves T lymphocytes. However, the mechanisms of T cell-mediated pathogenesis remain unknown. We found that, in contrast to ECM-susceptible C57BL6 mice, perforin-deficient (PFP-KO) mice were resistant to ECM in the absence of brain lesions, whereas cytoadherence of parasitized erythrocytes and massive accumulation of activated/effector CD8 lymphocytes were observed in both groups of mice. ECM is induced in PFP-KO mice after adoptive transfer of cytotoxic CD8+ cells from infected C57BL6 mice, which were directed to the brain of PFP-KO mice. This specific recruitment might involve chemokine/chemokine receptors, since their expression was up-regulated on activated CD8 cells, and susceptibility to ECM was delayed in CCR5-KO mice. Thus, lymphocyte cytotoxicity and cell trafficking are key players in ECM pathogenesis.

No full-text available

Request Full-text Paper PDF

To read the full-text of this research,
you can request a copy directly from the authors.

... 16,17 Mice deficient in granzyme B and perforin are completely protected against ECM reinforcing the belief that neurological disease is primarily a manifestation of cytotoxic T lymphocyte (CTL)-mediated tissue damage. [18][19][20] Some evidence indicates that CTL requires an additional antigenic stimulus locally within the brain in order to release their cytotoxic enzymes 18 and that endothelial cells (ECs) provide this stimulus via MHC class I-mediated parasite antigen presentation. 13 However, reporter mice that reveal professional antigen-presenting cell (APC) populations were not used in this in vivo study, despite previous reports showing that antigen-specific CD8 + T cells make long-lasting contact with CNS-resident CX 3 CR1 + APC in the perivascular space. ...
... This was consistent with the reported inability of these cells to reverse the resistance of RAG-1 À/À mice to neurological disease. 17,19 In contrast, all mice that received primed CD8 + T cells exhibited clinical signs of illness on day 7 p.i. and began to progress to NS (Figure 2c). We noted that none of the DPE-GFPxRAG-1 À/À mice that received primed CD8 + T cells had a profound impairment of motor responses typically seen in DPE-GFP mice during NS ( Figure 1b). ...
... Quantification by flow cytometry showed that, compared to UI mice, increased numbers of CD8 + T cells sequestered in the brains of DPE-GFPxRAG-1 À/À mice that had received primed CD8 + T cells, which is consistent with previous reports. 12,19 We also performed dose titration experiments using increasing numbers of adoptively transferred primed CD8 + T cells (0, 10 and 22 9 10 6 ) (Supplementary figure 1d). Increasing the number of cells to 22 9 10 6 accelerated the disease course by 1-2 days, and by day 7 p.i., all the mice had progressed in their disease course and were euthanised. ...
Article
Full-text available
Objectives The immunologic events that build up to the fatal neurological stage of experimental cerebral malaria (ECM) are incompletely understood. Here, we dissected the immune cell behaviour that occurs in the central nervous system (CNS) when Plasmodium berghei-ANKA (PbA)-infected mice show only minor clinical signs. Methods We used a 2-photon intravital microscopy brain-imaging model to study the spatio-temporal context of early immunological events in situ during ECM. Results We found that early in the disease course, antigen-specific CD8+ T cells come in contact and arrest on the endothelium of post-capillary venules. CD8+ T cells typically adhered adjacent to or in the near vicinity of perivascular macrophages (PVM) that line the post-capillary venules. Closer examination revealed that CD8+ T cells crawled along the inner vessel wall towards PVM that lay on the abluminal side of large post-capillary venules. T cells typically localised at ‘activity hotspots’ in large post-capillary venules characterized by activated morphology and clustering of PVM, increased abutting of post-capillary venules by PVM as well as augmented monocyte accumulation. In the later stage of infection when mice exhibited neurological signs, intravascular CD8+ T cells increased in number and changed their behaviour, actively crawling along the endothelium and displaying frequent, short-term interactions with the inner vessel wall at hotspots. Conclusion Our study suggests an active interplay between the innate and adaptive arms of the immune system across the blood-brain barrier (BBB) in early ECM, which may be the initiating event in the inflammatory cascade leading to BBB alteration and neuropathology.
... In the second step, a feature set is chosen such that it contains features that are uncorrelated to each other. As the feature N IFN B has the Many studies have implicated CD8 + T cells more commonly than CD4 + T cells in the pathogenesis of ECM [33,[134][135][136][137]. Therefore, out of the other strongly correlated pair is added to the final feature set. ...
... However, this proportion is comparable between the mice infected with KO sporozoites or WT sporozoites, even though both the groups show a reduced proportion as compared to the control mice. CD8 + T cells have been implicated in many studies in cerebral immunopathalogy [33,[135][136][137]. After reaching the brain, cytotoxic CD8 + T cells are exposed to parasite antigens cross-presented by the activated endothelial cells, thereby contributing to heightened inflammation and further disruption of bloodbrain barrier. ...
... Studies have highlighted that CD8 + T cells found in the brain during the infection are indeed responsible for cerebral immunopathology [139]. Additionally, PbANKA WT infection is known to cause a peak in the proportion of CD8 + T cells in leukocytes in the spleen, followed by a drastic reduction in the proportion around the day when infected mice show ECM symptoms [33,[135][136][137]. Likewise, the comparative analysis done here, also suggests the proportion of CD8 + T cells in the spleen as the next important player in causing cerebral Malaria. ...
Thesis
Cerebral Malaria is a complex neurological condition that results from interaction between the host and the Plasmodium parasite through the different phases of parasite's life-cycle. This interaction ranges from infection to immune response triggered in the host system. Various strains of the Plasmodium parasites are found to have difference in the severity of disease after infection. However, the precise factors defining the infectivity of Plasmodium parasites and the resulting disease outcome have not been completely identified so far. In the thesis, the Plasmodium berghei mouse model for Malaria is used to characterize the infection dynamics of Plasmodium berghei ANKA (wild-type) and a mutant strain that lacks a Plasmodium antigen PbmaLS_05. The mutant infection leads to lower parasitemia in red blood cells and less severe disease outcome in contrast to mice infected with the wild-type strain. Moreover, the mice infected by injecting PbmaLS_05(-) KO-infected red blood cells show reduced immune response in contrast to infection with PbmaLS_05(-) KO-sporozoites. By developing mathematical models describing various mechanisms of the infection and fitting them to experimental data; I find factors that influence the difference in disease progression seen between the two strains. Most strikingly, the KO strain shows a decreased ability to infect immature red blood cells that are usually a preferred target of the parasite. This altered property of infection limits parasite burden and affects disease progression. In addition to this, a statistical analysis of immune activation and immune response data from the KO or WT infected mice was done, which resulted in selecting major indicators of cerebral Malaria. The analysis showed that the number of CD8+ T cells accumulated in the brain, the reduced proportion of CD8+ T cells to lymphocytes in the spleen, the increased presence of Malaria specific CD8+ IFN-+ T and the secondary activation of CD8+ T cells due to the antigens cross-presented by infected red blood cells sequestered in the brain are the prominent distinguishing factors between the ECM causing PbANKA and non-ECM causing PbmaLS_05 (-) infections. An exploratory analysis of the liver-stage of infection and immune response highlighted that PbmaLS_05 may not have an important role to play in triggered immune response during the liver-stage of Malaria. However, its absence may lead to a small decrease in number of productive infections during the liver-stage, which must be further investigated. The antigen PbmaLS_05 can potentially aid in discovering the factors that influence the activation of immune responses and that might contribute to vaccine development and efficient parasite control.
... [16][17][18] Further studies elucidated the requirement of the CD8s to be cytotoxically functional for neuropathogenesis to occur. 16,17,19,20 As in the human disease, pRBCs and CD8 T cells are observed to interact with brain microvasculature, infiltrating the brain, and are strongly implicated in the neuropathology. 12 Activated CD8 T cells produce IFN-γ, which has been shown to stimulate mouse brain endothelial cells to increase cross-presentation of malaria antigens in vitro and in mouse brain microvessel explants. ...
... 16,18 The requirement for their activation and cytotoxic competence to establish disease has been confirmed as well. 19,20,56 The importance of the molecular interactions of CD8 T cells with endothelium in cerebral malaria pathology was recently highlighted by Riggle et al., 13 who demonstrated cytotoxic CD8s interacting with endothelium in post-mortem human brain samples of patients who died of cerebral malaria. This work and that from others also illustrates the remarkable similarity of CD8 localization in humans with cerebral malaria and mice with ECM. ...
Article
PD-1 checkpoint blockade is a promising immunotherapeutic approach for many solid tumors; however, this strategy is minimally effective for GBM patients as a monotherapy. One potential reason why α-PD-1 checkpoint blockade is ineffective is the tumor microenvironment and systemic derangements of the immune system which are prevalent in GBM patients. We have successfully modeled peripheral immune derangements in experimental murine GBM models. These include low peripheral blood CD4 T cell counts, reduced MHC class II expression on monocytes, and atrophy of primary immune organs in animals harboring gliomas. We therefore hypothesized that extended half-life IL-2, a potent cytokine which promotes the proliferation, differentiation, and killing activity of T cells, could overcome immune derangements and could potentially synergize with α-PD-1 checkpoint blockade therapy. In cohorts of C57Bl/6 mice, we supplemented α-PD-1 checkpoint blockade with multiple bio-engineered extended half-life IL-2 molecules to treat GL261 and CT2A orthotopic glioma models which are normally resistant to monotherapy. We observe a statistically significant increase in survival in GL261-bearing animals receiving the combination of extended half-life IL-2 reagents and αPD-1 therapy relative to control-treated mice. Some animals receiving this combination therapy had a reduction in tumor burden to levels below detection. These long-term survivors underwent a rechallenge experiment with a second inoculation of GL261-Luc and effectively cleared tumor without further therapeutic intervention. This intervention causes a significant shift in the immune profile, including the formation of mature T cell responses, as well as restoring the diminished peripheral immune cell counts in GL261-bearing animals. Finally, combining extended half-life IL-2 with α-PD-1 checkpoint blockade is effective independently of the CD8 T cell response. Together, these data suggest that combination immunotherapies employing extended half-life IL-2 fusion proteins are translational to the clinic and do not require the identification of tumor specific antigens for efficacy
... [16][17][18] Further studies elucidated the requirement of the CD8s to be cytotoxically functional for neuropathogenesis to occur. 16,17,19,20 As in the human disease, pRBCs and CD8 T cells are observed to interact with brain microvasculature, infiltrating the brain, and are strongly implicated in the neuropathology. 12 Activated CD8 T cells produce IFN-γ, which has been shown to stimulate mouse brain endothelial cells to increase cross-presentation of malaria antigens in vitro and in mouse brain microvessel explants. ...
... 16,18 The requirement for their activation and cytotoxic competence to establish disease has been confirmed as well. 19,20,56 The importance of the molecular interactions of CD8 T cells with endothelium in cerebral malaria pathology was recently highlighted by Riggle et al., 13 who demonstrated cytotoxic CD8s interacting with endothelium in post-mortem human brain samples of patients who died of cerebral malaria. This work and that from others also illustrates the remarkable similarity of CD8 localization in humans with cerebral malaria and mice with ECM. ...
Article
Cerebral malaria is the deadliest complication that can arise from Plasmodium infection. CD8 T cell engagement of brain vasculature is a putative mechanism of neuropathology in cerebral malaria. To define contributions of brain endothelial cell MHC class I antigen-presentation to CD8 T cells in establishing cerebral malaria pathology, we developed novel H-2Kb LoxP and H-2Db LoxP mice crossed with Cdh5-Cre mice to achieve targeted deletion of discrete class I molecules, specifically from brain endothelium. This strategy allowed us to avoid off-target effects on iron homeostasis and class-I-like molecules which are known to perturb Plasmodium infection. This is the first endothelial-specific ablation of individual class-I molecules enabling us to interrogate these molecular interactions. In these studies we interrogate human and mouse transcriptomics data to compare antigen presentation capacity during cerebral malaria. Using the Plasmodium berghei ANKA model of experimental cerebral malaria (ECM), we observe that H-2Kb and H-2Db class I molecules regulate distinct patterns of disease onset, CD8 T cell infiltration, targeted cell death, and regional blood-brain barrier (BBB) disruption. Strikingly, ablation of either molecule from brain endothelial cells resulted in reduced CD8 T cell activation, attenuated T cell interaction with brain vasculature, lessened targeted cell death, preserved BBB integrity, and prevented ECM and the death of the animal. We were able to show that these events were brain specific through the use of parabiosis and created the novel technique of dual small animal MRI to simultaneously scan conjoined parabionts during infection. These data demonstrate that interactions of CD8 T cells with discrete MHC class I molecules on brain endothelium differentially regulate development of ECM neuropathology. Therefore, targeting MHC class I interactions therapeutically may hold potential for treatment of cases of severe malaria.
... Cerebral malaria (CM), caused by Plasmodium falciparum (P.f) infection, is one of the most dangerous complications of malaria infection. 1,2 Disruption of the blood-brain barrier (BBB), cerebral edema, and neurological symptoms are hallmarks of CM. [3][4][5] Although a variety of immune cells are involved in the immunopathological damage during BBB breakdown, CD8 + T cell depletion 6,7 or functional disruption, 8,9 even just 1 day before the onset of the neurological symptoms, could completely abrogate this disease in a mouse model named experimental cerebral malaria (ECM). Parasite-specific CD8 + T cells mediate the destruction of brain microvascular endothelial cells (BMECs), which is directly responsible for the loss of BBB integrity observed in the ECM. ...
... Parasite-specific CD8 + T cells mediate the destruction of brain microvascular endothelial cells (BMECs), which is directly responsible for the loss of BBB integrity observed in the ECM. [8][9][10] Besides BMECs, brain parenchyma cells are also targets of brain-infiltrated CD8 + T cells and play an important role in brain inflammation and central nervous system (CNS) injury. 11,12 Although the pathogenesis of human CM is not identical to that of ECM, brain-infiltrated cytotoxic CD8 + T cells play a key role in the pathogenesis of human CM, such as BBB breakdown and CNS inflammation. ...
Article
Full-text available
Introduction Cerebral malaria (CM) is a lethal neuroinflammatory disease caused by Plasmodium infection. Immune cells and brain parenchyma cells contribute to the pathogenesis of CM. However, a systematic examination of the changes that occur in the brain parenchyma region during CM at the single‐cell resolution is still poorly studied. Aims To explore cell composition and CD8⁺ T cell infiltration, single‐cell RNA sequencing (scRNA‐seq) was performed on the brainstems of healthy and experimental cerebral malaria (ECM) mice. Then CD8⁺ T cell infiltration was confirmed by flow cytometry and immunofluorescence assays. Subsequently, the characteristics of the brain‐infiltrated CD8⁺ T cells were analyzed. Finally, the interactions between parenchyma cells and brain‐infiltrated CD8⁺ T cells were studied with an astrocytes‐CD8⁺ T cell cocultured model. Results The brainstem is the most severely damaged site during ECM. ScRNA‐seq revealed a large number of CD8⁺ T cells infiltrating into the brainstem in ECM mice. Brain‐infiltrated CD8⁺ T cells were highly activated according to scRNA‐seq, immunofluorescence, and flow cytometry assays. Further analysis found a subset of ki‐67⁺ CD8⁺ T cells that have a higher transcriptional level of genes related to T cell function, activation, and proliferation, suggesting that they were exposed to specific antigens presented by brain parenchyma cells. Brain‐infiltrated CD8⁺ T cells were the only prominent source of IFN‐γ in this single‐cell analysis. Astrocytes, which have a high interferon response, act as cross‐presenting cells to recruit and re‐activate brain‐infiltrated CD8⁺ T cells. We also found that brain‐infiltrated CD8⁺ T cells were highly expressed immune checkpoint molecule PD‐1, while parenchyma cells showed up‐regulation of PD‐L1 after infection. Conclusions These findings reveal a novel interaction between brain‐infiltrated CD8⁺ T cells and parenchyma cells in the ECM brainstem, suggesting that the PD‐1/PD‐L1 signal pathway is a promising adjunctive therapeutic strategy for ECM targeting over‐activated CD8⁺ T cells.
... We then imaged, as further controls, the iRBC distribution in PbA-infected perforin-deficient mice (C57BL/6 prf −/− ) (Fig. 1G-I) and BALB/c mice (Fig. 1J, K). In both C57BL/6 prf −/− and BALB/c mice PbA replicates in erythrocytes and causes anemia, but not ECM 26,27 . The spatial distribution of the iRBCs in C57BL/6 prf −/− mice closely mimicked that of the C57BL/6 wt mice (Fig. 1G-I), but the ratio of iRBCs in infected compared to non-infected mice was lower in C57BL/6 prf −/− mice (Fig. 1L). ...
Article
Full-text available
Sequestration of infected red blood cells (iRBCs) in the microcirculation is a hallmark of cerebral malaria (CM) in post-mortem human brains. It remains controversial how this might be linked to the different disease manifestations, in particular brain swelling leading to brain herniation and death. The main hypotheses focus on iRBC-triggered inflammation and mechanical obstruction of blood flow. Here, we test these hypotheses using murine models of experimental CM (ECM), SPECT-imaging of radiolabeled iRBCs and cerebral perfusion, MR-angiography, q-PCR, and immunohistochemistry. We show that iRBC accumulation and reduced flow precede inflammation. Unexpectedly, we find that iRBCs accumulate not only in the microcirculation but also in large draining veins and sinuses, particularly at the rostral confluence. We identify two parallel venous streams from the superior sagittal sinus that open into the rostral rhinal veins and are partially connected to infected skull bone marrow. The flow in these vessels is reduced early, and the spatial patterns of pathology correspond to venous drainage territories. Our data suggest that venous efflux reductions downstream of the microcirculation are causally linked to ECM pathology, and that the different spatiotemporal patterns of edema development in mice and humans could be related to anatomical differences in venous anatomy.
... In the brain, infected erythrocytes attach to the endothelium, leading to a further increase in chemokine and cytokine levels, which activate endothelial cells that uniquely perform cross-presentation of parasite antigens [23]. Activated CD8 + T cells from the periphery migrate to the brain, recognize Plasmodium antigens and kill the antigen-presenting endothelial cells by releasing perforin and granzyme B (GzmB) [24,25]. Destabilization of the blood-brain barrier facilitates an influx of peripheral immune cells into the brain [23]. ...
Article
Full-text available
Neuropilin-1 (Nrp-1) expression on CD8 ⁺ T cells has been identified in tumor-infiltrating lymphocytes and in persistent murine gamma-herpes virus infections, where it interferes with the development of long-lived memory T cell responses. In parasitic and acute viral infections, the role of Nrp-1 expression on CD8 ⁺ T cells remains unclear. Here, we demonstrate a strong induction of Nrp-1 expression on CD8 ⁺ T cells in Plasmodium berghei ANKA (PbA)-infected mice that correlated with neurological deficits of experimental cerebral malaria (ECM). Likewise, the frequency of Nrp-1 ⁺ CD8 ⁺ T cells was significantly elevated and correlated with liver damage in the acute phase of lymphocytic choriomeningitis virus (LCMV) infection. Transcriptomic and flow cytometric analyses revealed a highly activated phenotype of Nrp-1 ⁺ CD8 ⁺ T cells from infected mice. Correspondingly, in vitro experiments showed rapid induction of Nrp-1 expression on CD8 ⁺ T cells after stimulation in conjunction with increased expression of activation-associated molecules. Strikingly, T cell-specific Nrp-1 ablation resulted in reduced numbers of activated T cells in the brain of PbA-infected mice as well as in spleen and liver of LCMV-infected mice and alleviated the severity of ECM and LCMV-induced liver pathology. Mechanistically, we identified reduced blood-brain barrier leakage associated with reduced parasite sequestration in the brain of PbA-infected mice with T cell-specific Nrp-1 deficiency. In conclusion, Nrp-1 expression on CD8 ⁺ T cells represents a very early activation marker that exacerbates deleterious CD8 ⁺ T cell responses during both, parasitic PbA and acute LCMV infections.
... The hallmark of ECM is the accumulation and adherence of pathogenic CD8 + T cells in cerebral blood vessels [33][34][35]. In ECM, conventional CD11c + CD8α + DCs process and present parasitic antigens to CD8 + T cells, which subsequently contribute to the disruption of the blood-brain barrier (BBB) through granzyme B and perforin-mediated cytotoxicity [36][37][38][39]. ...
Article
Full-text available
The cytokine tumor necrosis factor (TNF) critically regulates the intertwined cell death and pro-inflammatory signaling pathways of dendritic cells (DCs) via ubiquitin modification of central effector molecules, but the intrinsic molecular switches deciding on either pathway are incompletely defined. Here, we uncover that the ovarian tumor deubiquitinating enzyme 7b (OTUD7b) prevents TNF-induced apoptosis of DCs in infection, resulting in efficient priming of pathogen-specific CD8+ T cells. Mechanistically, OTUD7b stabilizes the E3 ligase TNF-receptor-associated factor 2 (TRAF2) in human and murine DCs by counteracting its K48-ubiquitination and proteasomal degradation. TRAF2 in turn facilitates K63-linked polyubiquitination of RIPK1, which mediates activation of NF-κB and MAP kinases, IL-12 production, and expression of anti-apoptotic cFLIP and Bcl-xL. We show that mice with DC-specific OTUD7b-deficiency displayed DC apoptosis and a failure to induce CD8+ T cell-mediated brain pathology, experimental cerebral malaria, in a murine malaria infection model. Together, our data identify the deubiquitinating enzyme OTUD7b as a central molecular switch deciding on survival of human and murine DCs and provides a rationale to manipulate DC responses by targeting their ubiquitin network downstream of the TNF receptor pathway.
... The interaction of antigen-specific CD8 + T cells and central nervous system-residing APCs was associated with BBB damage and vascular leakage in the brain and meninges especially in olfactory bulb and brainstem [51]. CD8 + T cells show a cytotoxic function through production of perforin and granzyme B for development of ECM [60][61][62]. The evidence showed perforin from antigen-specific CD8 + T cells induced endothelial cell death [60], more importantly antigenspecific CD8 + T cells alter endothelial cell tight junctions that cause vascular leakage into the brain [51]. ...
... In the CNS, astrocytes and microglia are prominent sources of CXCL10 (IP-10; interferon gamma-induced protein 10) and CXCL9 (MIG; monokine induced by interferon gamma) [42]. IP-10 attracts activated T cells and mediates the Th1 response, which is characterized by the sequestration of monocytes and CD8 + T cells [43,44]. Granule exocytosis and Fas-FasL activation cause death of brain endothelial cells by CD8 + lymphocytes, which might result in BBB disruption [45]. ...
Article
Full-text available
Purpose: Here, we investigated expression modules reflecting the reciprocal expression of the cancer microenvironment and immune response-related genes associated with poor prognosis in primary central nervous system lymphoma (PCNSL). Methods: Weighted gene coexpression network analysis revealed representative modules, including neurogenesis, immune response, anti-virus, microenvironment, gene expression and translation, extracellular matrix, morphogenesis, and cell adhesion in the transcriptome data of 31 PCNSL samples. RESULTS : Gene expression networks were also reflected by protein-protein interaction networks. In particular, some of the hub genes were highly expressed in patients with PCNSL with prognoses as follows: AQP4, SLC1A3, GFAP, CXCL9, CXCL10, GBP2, IFI6, OAS2, IFIT3, DCN, LRP1, and LUM with good prognosis; and STAT1, IFITM3, GZMB, ISG15, LY6E, TGFB1, PLAUR, MMP4, FTH1, PLAU, CSF3R, FGR, POSTN, CCR7, TAS1R3, small ribosomal subunit genes, and collagen type 1/3/4/6 genes with poor prognosis. Furthermore, prognosis prediction formulae were constructed using the Cox proportional-hazards regression model, which demonstrated that the IP-10 receptor gene CXCR3 and type I interferon-induced protein gene IFI44L could predict patient survival in PCNSL. Conclusion: These results indicate that the differential expression and balance of immune response and microenvironment genes may be required for PCNSL tumor growth or prognosis prediction, which would help understanding the mechanism of tumorigenesis and potential therapeutic targets in PCNSL.
... It was also found that the elevated levels of caspase-3 and calpain1 are associated with disease pathology (Shukla et al. 2006) and inhibition of Caspase-3, and overexpression of Bcl-2 (an antiapoptotic protein) could prevent ECM (Helmers et al. 2008). Mice that harbour T cells lacking perforin Nitcheu et al. 2003) and Granzyme-b (Gra-b) (Haque et al. 2011) were protected from ECM, indicating the critical role of caspase-3 and T cells in ECM pathogenesis. Furthermore, Lymphotoxin-α inhibition has protected the mice from ECM by decreasing complement receptor, CR3, and cell adhesion molecule ICAM-1 (Engwerda et al. 2002), implying that Lt-α is a crucial regulator or all the factors involved in ECM pathogenesis associate with Lymphotoxin-α. ...
Article
Full-text available
Knockdown studies have shown lymphotoxin-α (Lt-α) as a critical molecule for Experimental cerebral malaria (ECM) pathogenesis. We investigated the role of lymphotoxin-α in regulating active caspase-3 and calpain1. T cell infiltration into the brains, and subsequent neuronal cell death are the essential features of Plasmodium berghei ANKA(PbA)-induced ECM. Our results showed increased Lt-α levels during ECM. Treatment of naïve mice with serum from ECM mice and exogenous Lt-α was lethal. We inhibited Lt-α in vivo during PbA infection by injecting the mice with anti-Lt-α antibody. Inhibition of Lt-α mitigated neuronal cell death and increased mice’s survival until 30-day post-infection (p.i.) compared to only 15 days survival of PbA control mice.
... The experimental murine model of cerebral malaria (ECM) has highlighted the central role of CD8 T cells in ECM development through their implication in endothelial cell apoptosis (Nitcheu et al., 2003;Howland et al., 2015). The implication of monocytes/macrophages in the pathophysiology of CM has also been evidenced both in humans (Pongponratn et al., 2003;Stanisic et al., 2014) and in a murine model, where these cells have been shown to accumulate in the brain microvasculature at the onset of neurological symptoms (Niewold et al., 2018;Sierro and Grau, 2019). ...
Article
Full-text available
Cerebral malaria (CM) is one of the most severe forms of malaria and is a neuropathology that can lead to death. Monocytes have been shown to accumulate in the brain microvasculature at the onset of neurological symptoms during CM. Monocytes have a remarkable ability to adapt their function to their microenvironment from pro-inflammatory to resolving activities. This study aimed to describe the behavior of monocyte subpopulations during infection and its resolution. C57BL/6 mice were infected with the Plasmodium berghei ANKA strain and treated or not with chloroquine (CQ) on the first day of the onset of neurological symptoms (day 6) for 4 days and followed until day 12 to mimic neuroinflammation and its resolution during experimental CM. Ly6C monocyte subpopulations were identified by flow cytometry of cells from the spleen, peripheral blood, and brain and then quantified and characterized at different time points. In the brain, the Ly6Cint and Ly6Clow monocytes were associated with neuroinflammation, while Ly6Chi and Ly6Cint were mobilized from the peripheral blood to the brain for resolution. During neuroinflammation, CD36 and CD163 were both involved via splenic monocytes, whereas our results suggest that the low CD36 expression in the brain during the neuroinflammation phase was due to degradation. The resolution phase was characterized by increased expressions of CD36 and CD163 in blood Ly6Clow monocytes, a higher expression of CD36 in the microglia, and restored high expression levels of CD163 in Ly6Chi monocytes localized in the brain. Thus, our results suggest that increasing the expressions of CD36 and CD163 specifically in the brain during the neuroinflammatory phase contributes to its resolution.
... The experimental murine model of cerebral malaria (ECM) has highlighted the central role of CD8 T cells in ECM development through their implication in endothelial cell apoptosis (Nitcheu et al., 2003;Howland et al., 2015). The implication of monocytes/macrophages in the pathophysiology of CM has also been evidenced both in humans (Pongponratn et al., 2003;Stanisic et al., 2014) and in a murine model, where these cells have been shown to accumulate in the brain microvasculature at the onset of neurological symptoms (Niewold et al., 2018;Sierro and Grau, 2019). ...
... It was also found that the elevated levels of caspase-3 and calpain1 are associated with disease pathology [21] and inhibition of Caspase-3, and overexpression of Bcl-2 (an anti-apoptotic protein) could prevent ECM [22]. Mice that harbour T cells lacking perforin [23,24] and Granzyme-b (Gra-b) [25] were protected from ECM, indicating the critical role of caspase-3 and T cells in ECM pathogenesis. Further, Lymphotoxin-α inhibition has protected the mice from ECM by decreasing complement receptor, CR3, and cell adhesion molecule ICAM-1 [26], implying that Lt-α is a crucial regulator or all the factors involved in ECM pathogenesis associate with Lymphotoxin-α. ...
Preprint
Full-text available
Knockdown studies have shown lymphotoxin-α (Lt-α) as a critical molecule for Experimental cerebral malaria (ECM) pathogenesis. We investigated the role of lymphotoxin-α in regulating active caspase-3 and calpain1. T cell infiltration into the brains, and subsequent neuronal cell death are the essential features of Plasmodium berghei ANKA(PbA) induced ECM in C57BL/6 mice. Our results showed increased Lt-α levels during ECM. Treatment of naïve mice with serum from ECM mice and exogenous Lt-α was lethal. We inhibited Lt-α in vivo during PbA infection by injecting the mice with anti-Lt-α antibody. Inhibition of Lt-α mitigated neuronal cell death and increased mice's survival until 30 days post-infection (p.i) compared to only 15 days survival of PbA control mice.
... The interaction of antigen-specific CD8 + T cells and central nervous system-residing APCs was associated with BBB damage and vascular leakage in the brain and meninges especially in olfactory bulb and brainstem [51]. CD8 + T cells show a cytotoxic function through production of perforin and granzyme B for development of ECM [60][61][62]. The evidence showed perforin from antigen-specific CD8 + T cells induced endothelial cell death [60], more importantly antigenspecific CD8 + T cells alter endothelial cell tight junctions that cause vascular leakage into the brain [51]. ...
Article
Malaria is a life-threatening disease caused by infection with Plasmodium parasites. The goal of developing an effective malaria vaccine is yet to be reached despite decades of massive research efforts. CD4⁺ helper T cells, CD8⁺ cytotoxic T cells, and γδ T cells are associated with immune responses to both liver-stage and blood-stage Plasmodium infection. The immune responses of T cell-lineages to Plasmodium infection are associated with both protection and immunopathology. Studies with mouse model of malaria contribute to our understanding of host immune response. In this paper, we focus primarily on mouse malaria model with blood-stage Plasmodium berghei infection and review our knowledge of T cell immune responses against Plasmodium infection. Moreover, we also discuss findings of experimental human studies. Uncovering the precise mechanisms of T cell-mediated immunity to Plasmodium infection can be accomplished through further investigations using mouse models of malaria with rodent Plasmodium parasites. Those findings would be invaluable to advance the efforts for development of an effective malaria vaccine.
... It was also found that the elevated levels of caspase-3 and calpain1 are associated with disease pathology [21] and inhibition of Caspase-3, and overexpression of Bcl-2 (an anti-apoptotic protein) could prevent ECM [22]. Mice that harbour T cells lacking perforin [23,24] and Granzyme-b (Gra-b) [25] were protected from ECM, indicating the critical role of caspase-3 and T cells in ECM pathogenesis. Further, Lymphotoxin-α inhibition has protected the mice from ECM by decreasing complement receptor, CR3, and cell adhesion molecule ICAM-1 [26], implying that Lt-α is a crucial regulator or all the factors involved in ECM pathogenesis associate with Lymphotoxin-α. ...
Preprint
Full-text available
Knockdown studies have shown lymphotoxin-α (Lt-α) as a critical molecule for Experimental cerebral malaria (ECM) pathogenesis. We investigated the role of lymphotoxin-α in regulating active caspase-3 and calpain1. T cell infiltration into the brains, and subsequent neuronal cell death are the essential features of Plasmodium berghei ANKA(PbA) induced ECM in C57BL/6 mice. Our results showed increased Lt-α levels during ECM. Treatment of naïve mice with serum from ECM mice and exogenous Lt-α was lethal. We inhibited Lt-α in vivo during PbA infection by injecting the mice with anti-Lt-α antibody. Inhibition of Lt-α mitigated neuronal cell death and increased mice's survival until 30 days post-infection (p.i) compared to only 15 days survival of PbA control mice.
... T cells accumulation in brain microvessels plays a vital role in cerebral malaria pathogenesis [58]. The roles of CD4 + [55] and CD8 + [58,59] cells in disease assistance have been determined experimentally. Studies suggested that the sequestration of leukocytes and parasitized RBC in brain microvessels helps in disease induction [60,61]. ...
Article
Full-text available
The severe form of malaria, i.e., cerebral malaria caused by Plasmodium falciparum, is a complex neurological syndrome. Surviving persons have a risk of behavioral difficulties, cognitive disorders, and epilepsy. Cerebral malaria is associated with multiple organ dysfunctions. The adhesion and accumulation of infected RBCs, platelets, and leucocytes (macrophages, CD4+ and CD8+ T cells, and monocytes) in the brain microvessels play an essential role in disease progression. Micro-vascular hindrance by coagulation and endothelial dysfunction contributes to neurological damage and the severity of the disease. Recent studies in human cerebral malaria and the murine model of cerebral malaria indicate that different pathogens as well as host-derived factors are involved in brain microvessel adhesion and coagulation that induces changes in vascular permeability and impairment of the blood-brain barrier. Efforts to alleviate blood-brain barrier dysfunction and de-sequestering of RBCs could serve as adjunct therapies. In this review, we briefly summarize the current understanding of the pathogenesis of cerebral malaria, the role of some factors (NK cells, platelet, ANG-2/ANG-1 ratio, and PfEMP1) in disease progression and various functions of Mesenchymal stem cells. This review also highlighted the implications of MSCs as a regenerative medicine.
... In mouse models of experimental cerebral malaria (ECM) caused by Plasmodium berghei, inflammatory responses regulated by transcription factors, cytokines and signaling pathways that mediate pro-inflammatory responses are required for pathology (Longley et al., 2011), which is largely driven by T cells (Nitcheu et al., 2003;Villegas-Mendez et al., 2012). Before 2000, classical genetic studies of cerebral malaria were complicated due to the severe susceptibility of most laboratory mouse strains to the P. berghei ANKA strain. ...
Article
Full-text available
Forward genetic approaches have been widely used in parasitology and have proven their power to reveal the complexities of host-parasite interactions in an unbiased fashion. Many aspects of the parasite’s biology, including the identification of virulence factors, replication determinants, antibiotic resistance genes, and other factors required for parasitic life, have been discovered using such strategies. Forward genetic approaches have also been employed to understand host resistance mechanisms to parasitic infection. Here, we will introduce and review all forward genetic approaches that have been used to identify host factors involved with Apicomplexa infections, which include classical genetic screens and QTL mapping, GWAS, ENU mutagenesis, overexpression, RNAi and CRISPR-Cas9 library screens. Collectively, these screens have improved our understanding of host resistance mechanisms, immune regulation, vaccine and drug designs for Apicomplexa parasites. We will also discuss how recent advances in molecular genetics give present opportunities to further explore host-parasite relationships.
... Additionally, sequestered CD8+ T cells showed positive staining for the cytolytic protease granzyme B (GrB) (63). As enzymes such as GrB (66) and perforin (67) are responsible for apoptosis of endothelial cells in ECM, the findings presented by Riggle et al. indicate a similar mechanism in human CM and underline the relevance of the murine malaria model. ...
Article
Full-text available
This review focuses on current clinical and immunological aspects of cerebral malaria induced by Plasmodium falciparum infection. Albeit many issues concerning the inflammatory responses remain unresolved and need further investigations, current knowledge of the underlying molecular mechanisms is highlighted. Furthermore, and in the light of significant limitations in preventative diagnosis and treatment of cerebral malaria, this review mainly discusses our understanding of immune mechanisms in the light of the most recent research findings. Remarkably, the newly proposed CD8+ T cell-driven pathophysiological aspects within the central nervous system are summarized, giving first rational insights into encouraging studies with immune-modulating adjunctive therapies that protect from symptomatic cerebral participation of Plasmodium falciparum infection.
... Although CD36 is one receptor P. berghei infected erythrocytes interact with, the possibility of other, yet undefined, receptor-ligand interactions cannot be excluded. For example, cerebral malaria (CM) caused by P. falciparum is the result of sequestration of infected erythrocytes in the host microvascular (MacPherson et al., 1985), but experimental cerebral malaria (eCM) caused by P. berghei infection is thought to be due to congestion caused by immune cell infiltrate, even though the outward disease present similarly to CM (Chen et al., 2000;Hearn et al., 2000;Belnoue et al., 2002;Nitcheu et al., 2003). Indeed, P. berghei infection in CD36-/-mice still resulted in cases of eCM . ...
Thesis
Ein Kennzeichen der Plasmodium-Infektion ist die Induktion von strukturellen Veränderungen der Membranen der Wirtszelle. Die Parasiten bilden sich Membranstrukturen im Zytoplasma der Wirtszelle aus. Die Maurersche Fleckung ist einer der am besten charakterisierten Struktur von Plasmodium falciparum, die für den Transport von Virulenzfaktoren an die Oberfläche der infizierten Erythrozytenmembran wichtig ist. Plasmodium berghei, eine Nagetier-infizierende Spezies, bildet ähnliche Strukturen aus, die als intra-erythrozytäre P. berghei induzierte Strukturen (IBIS) bezeichnet werden. In beiden Strukturen können Proteine gefunden werden, die für die Sequestrierung von infizierten roten Blutkörperchen innerhalb des Wirts verantwortlich sind. Zwei P. berghei Proteine, IPIS2 und IPIS3, sind im Leber- und Blutstadium der Infektion präsent. Im Blutstadium werden sie in die IBIS-Kompartimente exportiert. Das speziesübergreifende Vorkommen von IPIS2 und IPIS3 deutet darauf hin, dass sie konservierte Funktionen haben könnten, die für das intrazelluläre Wachstum von Plasmodium wichtig sind. Diese Arbeit beschreibt Rollen für beide Proteine an der Wirts-Parasit Schnittstelle während der Infektion. Um einen Einblick in die induzierten Veränderungen zu erhalten, wurde die Infektion von P. berghei in Abwesenheit von IPIS2 beziehungsweise IPIS3 charakterisiert. Darüber hinaus interessierte uns auch, wie sich die Orthologe von IPIS2- und IPIS3 in den Blutstadien von P. knowlesi verhalten. Darüber hinaus zeigt diese Arbeit, dass die Deletion von entweder IPIS2 oder IPIS3 zu leicht reduzierten Wachstumsraten der Parasiten im Blut infizierter Tiere führte und die Anzahl der zirkulierenden Schizonten im peripheren Blut erhöhte, während das Leberstadium unbeeinflusst blieb. Zusammengenommen deuten diese Daten darauf hin, dass IPIS2 und IPIS3 für den Wirtszell-Umbau erforderlich sind, der für die effiziente Sequestrierung von infizierten Erythrozyten aus dem Blutkreislauf verantwortlich ist.
... Il semblerait que ces cellules ne soient pas directement impliquées dans le développement d'un ECM et joueraient un rôle mineur dans le recrutement des lymphocytes au cerveau (233,481,482). En revanche l'accumulation des LT et principalement des LT CD8 cytotoxiques a été largement décrite dans le cerveau des souris infectées et est associée à l'altération de la BHE et au décès des souris (124,125,330,481,485). Très récemment, ce phénomène a aussi été observé dans le cerveau de patients atteints de neuropaludisme (464,486). ...
Thesis
En 2020, le paludisme reste un problème majeur de santé publique. Malgré des avancées scientifiques capitales, il est encore responsable de plus de 400 000 décès chaque année, principalement chez les enfants en Afrique Sub-saharienne. Le neuropaludisme est l'une des formes les plus sévères du paludisme. Sa physiopathologie est complexe, impliquant l'adhésion des érythrocytes infectés par Plasmodium falciparum à l'endothélium vasculaire dans le cerveau. Ce mécanisme conduit à une obstruction du flux sanguin, une inflammation locale, une altération de la barrière hémato-encéphalique et à un éventail de réponses cellulaires visant à résoudre le processus de neuroinflammation. Parmi ces cellules, les monocytes/macrophages jouent un rôle clé de par leur capacité à s'adapter à leur microenvironnement en fonction des différents signaux qu'ils perçoivent, modulant ainsi les balances pro/anti-inflammatoire et pro/antioxydante, essentielles à la résolution du neuropaludisme. Les monocytes sont composés de trois sous-populations dont le rôle dans la physiopathologie du neuropaludisme a été peu étudié et reste à éclaircir. Pour compléter ces connaissances, une approche combinant modèle murin et études chez des patients béninois a été menée. Une première étude a été conduite chez des enfants béninois présentant un paludisme simple, une anémie sévère palustre, ou un neuropaludisme dans le but de déterminer si les paramètres monocytaires constituent des facteurs de sévérité et/ou de risque de décès au cours d'un paludisme grave. Les pourcentages et phénotypes des sous-populations monocytaires obtenus chez les trois groupes d'enfants lors de leur admission dans les centres de santé (J0) ont été comparés. Les résultats ont permis de mettre en lumière l'implication des monocytes non classiques dans la protection vis-à-vis des formes sévères de paludisme et de confirmer l'importance du récepteur CD36 des monocytes comme facteur protecteur de sévérité et de survenue de décès. Pour aller plus loin, un travail a ensuite été mené sur un modèle murin de neuropaludisme mis en place pour se rapprocher des conditions réelles, à savoir un traitement tardif, et débouchant sur une résolution de l'infection. Des souris C57BL/6 ont été infectées par P. berghei ANKA (J0) et traitées tardivement à la chloroquine. La cinétique d'évolution des sous-populations monocytaires a été suivie en termes de pourcentage et d'expression protéique et génique, à la fois dans le sang, la rate et le cerveau, jusqu'à la résolution de l'infection (J12). Les résultats suggèrent une implication des monocytes classiques et intermédiaires dans la neuroinflammation, au contraire des monocytes non classiques. A J12 était observé un nouvel afflux de monocytes intermédiaires vers le cerveau, suggérant également un rôle de cette sous-population dans la résolution tardive de la neuroinflammation. Les analyses protéique et génique ont mis en évidence une implication favorable de Nrf2, CD36, HO-1, CMH-II, COX-2 et de la 12/15-LOX dans la résolution de l'infection. Une deuxième étude a ensuite été menée chez des enfants béninois, avec un plus grand nombre de patients et un suivi à J3 et J30. Deux groupes de sujets ont été inclus, présentant un paludisme simple (n = 94) ou un neuropaludisme (n = 65). Dans l'ensemble, les résultats indiquent que les monocytes non classiques et intermédiaires étaient respectivement impliqués dans l'amélioration clinique et la survie des patients. L'expression de CD14, CD16, CD36 et HLA-DR était un facteur protecteur vis-à-vis du neuropaludisme. De manière intéressante, nos résultats montrent également une association entre une réponse pro-oxydante moins active, la sévérité du paludisme, et la survenue de décès. L'ensemble de ces résultats apporte un nouvel éclairage sur l'implication des monocytes/macrophages au cours du neuropaludisme.
... Similarly as in experimental MA-ARDS, CD8 + T cells play an important pathogenic role in experimental cerebral malaria (ECM) [48]. ECM pathology is dependent on perforin and granzyme B, demonstrating the important contribution of these cytotoxic markers to pathology [49,50]. Recently, Riggle et al. observed that CM in children was associated with cerebrovascular engagement of CD8 + T cells expressing granzyme B [51]. ...
Article
Full-text available
Malaria is a hazardous disease caused by Plasmodium parasites and often results in lethal complications, including malaria-associated acute respiratory distress syndrome (MA-ARDS). Parasite sequestration in the microvasculature is often observed, but its role in malaria pathogenesis and complications is still incompletely understood. We used skeleton binding protein-1 (SBP-1) KO parasites to study the role of sequestration in experimental MA-ARDS. The sequestration-deficiency of these SBP-1 KO parasites was confirmed with bioluminescence imaging and by measuring parasite accumulation in the lungs with RT-qPCR. The SBP-1 KO parasites induced similar lung pathology in the early stage of experimental MA-ARDS compared to wildtype (WT) parasites. Strikingly, the lung pathology resolved subsequently in more than 60% of the SBP-1 KO infected mice, resulting in prolonged survival despite the continuous presence of the parasite. This spontaneous disease resolution was associated with decreased inflammatory cytokine expression measured by RT-qPCR and lower expression of cytotoxic markers in pathogenic CD8 ⁺ T cells in the lungs of SBP-1 KO infected mice. These data suggest that SBP-1-mediated parasite sequestration and subsequent high parasite load are not essential for the development of experimental MA-ARDS but inhibit the resolution of the disease.
... In mice, ECM pathology has been strongly associated to effector CD8 + T cells primed in the spleen that infiltrate the brain (52,53). The induction of Plasmodium-specific T cells in infected WT mice towards antigens from different life cycle stages of PbA parasites has been shown before (54,55). ...
Article
Full-text available
Cerebral malaria is a potentially lethal disease, which is caused by excessive inflammatory responses to Plasmodium parasites. Here we use a newly developed transgenic Plasmodium berghei ANKA (PbAAma1OVA) parasite that can be used to study parasite-specific T cell responses. Our present study demonstrates that Ifnar1-/- mice, which lack type I interferon receptor-dependent signaling, are protected from experimental cerebral malaria (ECM) when infected with this novel parasite. Although CD8⁺ T cell responses generated in the spleen are essential for the development of ECM, we measured comparable parasite-specific cytotoxic T cell responses in ECM-protected Ifnar1-/- mice and wild type mice suffering from ECM. Importantly, CD8⁺ T cells were increased in the spleens of ECM-protected Ifnar1-/- mice and the blood-brain-barrier remained intact. This was associated with elevated splenic levels of CCL5, a T cell and eosinophil chemotactic chemokine, which was mainly produced by eosinophils, and an increase in eosinophil numbers. Depletion of eosinophils enhanced CD8⁺ T cell infiltration into the brain and increased ECM induction in PbAAma1OVA-infected Ifnar1-/- mice. However, eosinophil-depletion did not reduce the CD8⁺ T cell population in the spleen or reduce splenic CCL5 concentrations. Our study demonstrates that eosinophils impact CD8⁺ T cell migration and proliferation during PbAAma1OVA-infection in Ifnar1-/- mice and thereby are contributing to the protection from ECM.
... The activated CD8 + T cells then migrate to the brain in a CXCR3-CXCL10dependent manner and form long-lasting cognate interactions with parasite antigen cross-presenting endothelial cells, with CXCL10 playing an additional pathological role in the brain by stabilizing the binding of CD8 + T cells with the endothelial cells [9][10][11][12][13][14]. IFN-γ plays an important role in ECM pathogenesis by promoting endothelial cell cross presentation, MHC-class I expression and CXCL10 production within the brain, facilitating CD8 + T-cell entry and retention in the brain [10,11,15,16]. Following interaction with parasite-cross-presenting endothelial cells, CD8 + T cells mediate disruption of the blood brain barrier through Perforin and Granzyme B (GrB) production [17][18][19]. Notably, potential roles for CD8 + T cells in HCM has recently been described, with CD8 + T cells found in similar numbers in the brains during fatal HCM as is observed in ECM [20,21]. Moreover, CD8 + T cells appear to degranulate and release GrB proximal with brain endothelial cells in HCM [21], and the proportions of CD8 + T cells producing GrB were significantly higher in children with severe malaria than in those with uncomplicated malaria [22]. ...
Article
Full-text available
Experimental cerebral malaria (ECM) is a severe complication of Plasmodium berghei ANKA (PbA) infection in mice, characterised 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 are 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.
... During ECM pathogenesis, the activated endothelial cells adhere CD8 + T cells and present parasite-specific antigens to T cells. The effector CD8 + T cells secrete granzyme B and perforin to induce damage of intercellular tight junctions of the endothelium, trigger apoptosis of endothelial cells and neuronal cells, which disrupting the BBB and finally impairing central nervous system function (36)(37)(38). Thus, we measured the expression of granzyme B and perforin in the brain 7 dpi. ...
Article
Full-text available
Reticulon and the REEP family of proteins stabilize the high curvature of endoplasmic reticulum tubules. The REEP5 homolog in Plasmodium, Plasmodium berghei YOP1 (PbYOP1), plays an important role in the erythrocytic cycle of the P. berghei ANKA and the pathogenesis of experimental cerebral malaria (ECM), but the mechanisms are largely unknown. Here, we show that protection from ECM in Pbyop1Δ-infected mice is associated with reduced intracerebral Th1 accumulation, decreased expression of pro-inflammatory cytokines and chemokines, and attenuated pathologies in the brainstem, though the total number of CD4⁺ and CD8⁺ T cells sequestered in the brain are not reduced. Expression of adhesive molecules on brain endothelial cells, including ICAM-1, VCAM-1, and CD36, are decreased, particularly in the brainstem, where fatal pathology is always induced during ECM. Subsequently, CD8⁺ T cell-mediated cell apoptosis in the brain is compromised. These findings suggest that Pbyop1Δ parasites can be a useful tool for mechanistic investigation of cerebral malaria pathogenesis.
... DCs exposed to sHZ exhibited impaired PMAelicited ROS production, an effect exacerbated in CLEC12A -/-DCs, but pro-inflammatory cytokine and activation marker expression were not affected by the presence or absence of CLEC12A. Given the oft-cited role for activated CD8+ T cells in driving experimental CM [88][89][90], activation of CD8+ T cells exposed to intact and CLEC12A -/-sHZ-stimulated DCs was assessed. Lack of CLEC12A on DCs attenuated proinflammatory and cytotoxic molecule secretion by CD8+ T cells. ...
Article
Full-text available
Purpose of Review Malaria is a disease caused by parasites that reside in host red blood cells and use hemoglobin as a nutrient source. Heme released by hemoglobin catabolism is modified by the parasite to produce hemozoin (HZ), which has toxic effects on the host. Experimentation aiming to elucidate how HZ contributes to malaria pathogenesis has utilized different preparations of this molecule, complicating interpretation and comparison of findings. We examine natural synthesis and isolation of HZ and highlight studies that have used multiple preparations, including synthetic forms, in a comparative fashion. Recent Findings Recent work utilizing sophisticated imaging and detection techniques reveals important molecular characteristics of HZ synthesis and biochemistry. Other recent studies further refine understanding of contributions of HZ to malaria pathogenesis yet highlight the continuing need to characterize HZ preparations and contextualize experimental conditions in the in vivo infection milieu. Summary This review highlights the necessity of collectively determining what is physiologically relevant HZ. Characterization of isolated natural HZ and use of multiple preparations in each study are recommended with application of in vivo studies whenever possible. Adoption of such practices is expected to improve reproducibility of results and elucidate the myriad of ways that HZ participates in malaria pathogenesis.
... OVA-PbA (2,4,9,10), for which widely used murine T cell lines such as OT-I and OT-II 14 could be used to monitor specific T cell responses. While the use of these parasites in 15 conjunction with model T cell lines has aided the study of anti-malarial CD4 + and CD8 + T 16 cell responses (6,(11)(12)(13)(14)(15), wild-type parasites and transgenic T cells capable of recognizing 17 authentic parasite-derived antigens are preferred as they more closely resemble 18 endogenous responses to natural infections. With this in mind, we recently generated a 19 ...
Preprint
Full-text available
We describe an MHC II (IA b )-restricted T cell receptor (TCR) transgenic mouse line that produces CD4 ⁺ T cells specific for Plasmodium species. This line, termed PbT-II, was derived from a CD4 ⁺ T cell hybridoma generated to blood-stage Plasmodium berghei ANKA (PbA). PbT-II cells responded to all Plasmodium species and stages tested so far, including rodent (PbA, P. berghei NK65, P. chabaudi AS and P. yoelii 17XNL) and human ( P . falciparum) blood-stage parasites as well as irradiated PbA sporozoites. PbT-II cells can provide help for generation of antibody to P. chabaudi infection and can control this otherwise lethal infection in CD40L-deficient mice. PbT-II cells can also provide help for development of CD8 ⁺ T cell-mediated experimental cerebral malaria (ECM) during PbA infection. Using PbT-II CD4+ T cells and the previously described PbT-I CD8 ⁺ T cells, we determined the dendritic cell (DC) subsets responsible for immunity to PbA blood-stage infection. CD8 ⁺ DC (a subset of XCR1 ⁺ DC) were the major antigen presenting cell (APC) responsible for activation of both T cell subsets, though other DC also contributed to CD4 ⁺ T cell responses. Depletion of CD8 ⁺ DC at the beginning of infection prevented ECM development and impaired both Th1 and Tfh responses; in contrast, late depletion did not affect ECM. This study describes a novel and versatile tool for examining CD4 ⁺ T cell immunity during malaria and provides evidence that CD4 ⁺ T cell help, acting via CD40L signalling, can promote immunity or pathology to blood stage malaria largely through antigen presentation by CD8 ⁺ DC.
... As a side note, since viral control in ECs is linked to low levels of IL-15 in LNs , the usage of IL-15 superagonist as a cure strategy should be reassessed. On the other hand, deleterious effects of CD8+ T cells have been observed in several parasitic and viral infections, including leishmaniasis, malaria, Chagas disease, Coxsackie virus, and Zika virus infections (Ferreira et al., 2017;Henke, Huber, Stelzner, & Whitton, 1995;Jurado et al., 2018;Nitcheu et al., 2003;Novais et al., 2017;Novais et al., 2013). In these diseases, aberrant cytotoxicity mediated by pathogen-specific CD8+ T cells in tissues is linked to pathology. ...
Article
Acquired Immunodeficiency Syndrome (AIDS) is an immunodeficiency caused by infection with Human Immunodeficiency Virus (HIV). Since its first documentation in 1981, the AIDS pandemic has claimed the lives of 32 million people worldwide. The introduction of antiretroviral therapy drugs (ART) and combined therapy in 1995 has transformed HIV infections from a “death-sentence” into a manageable and often non-fatal chronic condition. Still, no cure is available. While without ART most HIV-infected individuals (>99%) progress to AIDS, a subset of infected individuals (
... CD8+ T cells can cause significant damage to tissues by secreting pro-inflammatory cytokines and/or by direct killing of target cells. Pathogenic effects of CD8+ T cells have been demonstrated in several parasitic and viral infections, including Leishmaniasis, Malaria, Chagas disease, Coxsackie virus, and Zika virus infections (Ferreira et al., 2017;Henke et al., 1995;Jurado et al., 2018;Nitcheu et al., 2003;Novais et al., 2017Novais et al., , 2013. In each of these diseases, the observed pathology has been attributed largely to CD8+ T cells with aberrant pathogenspecific perforin-mediated cytotoxic activity in the target tissues. ...
Article
CD8+ T cells are crucial for immunity against viral infections, including HIV. Several characteristics of CD8+ T cells, such as polyfunctionality and cytotoxicity, have been correlated with effective control of HIV. However, most of these correlates have been established in the peripheral blood. Meanwhile, HIV primarily replicates in lymphoid tissues. Therefore, it is unclear which aspects of CD8+ T cell biology are shared and which are different between blood and lymphoid tissues in the context of HIV infection. In this review, we will recapitulate the latest advancements of our knowledge on lymphoid tissue CD8+ T cells during HIV infection and discuss the insights these advancements might provide for the development of a HIV cure.
... Previous studies have shown that perforin-deficient mice are sensitive to obesity-induced insulin resistance as a result of restricted T cell expansion and activation in adipose tissue. Perforin has also been reported to play critical roles in promoting inflammation-mediated diseases, including type 1 diabetes (33), cerebral malaria (34), and viral myocarditis (35). A recent study revealed that perforin expressed in CD8 T cells regulates innate and adaptive immunity in the liver and exerts a protective effect in MCD (methionine/choline-deficient diet) diet-induced NASH models (36). ...
Article
Full-text available
Non-alcoholic fatty liver disease (NAFLD) is one of the main causes of cirrhosis and major risk factors for hepatocellular carcinoma and liver-related death. Despite substantial clinical and basic research, the pathogenesis of obesity-related NAFLD remains poorly understood. In this study, we show that perforin can act as an immune regulator to prevent the progression of NAFLD. Aged perforin-deficient (Prf−/−) mice have increased lipid accumulation in the liver compared to WT mice. With high-fat diet (HFD) challenge, Prf−/− mice have increased liver weight, more severe liver damage, and increased liver inflammation when compared with WT controls. Mechanistic studies revealed that perforin specifically regulates intrinsic IFN-γ production in CD4 T cells, not CD8 T cells. We found that CD4 T cell depletion reduces liver injury and ameliorates the inflammation and metabolic morbidities in Prf−/− mice. Furthermore, improved liver characteristics in HFD Prf−/− and IFN-γR−/− double knockout mice confirmed that IFN-γ is a key factor for mediating perforin regulation of NAFLD progression. Overall, our findings reveal the important regulatory role perforin plays in the progression of obesity-related NAFLD and highlight novel strategies for treating NAFLD.
... Most data on CD8 + T cells in blood-stage malaria are derived from murine models addressing their function in experimental cerebral malaria (ECM) using P. berghei ANKA infection of C57BL/6 mice (8). In murine malaria it was clearly shown that CD8 + T cells sequester in the brain and mediate endothelial leakage in a granzyme B (GrzB) and perforindependent cytolytic reaction (9)(10)(11)(12). ...
Article
Full-text available
In Plasmodium falciparum malaria, CD8⁺ T cells play a double-edged role. Liver-stage specific CD8⁺ T cells can confer protection, as has been shown in several vaccine studies. Blood-stage specific CD8⁺ T cells, on the other hand, contribute to the development of cerebral malaria in murine models of malaria. The role of CD8⁺ T cells in humans during the blood-stage of P. falciparum remains unclear. As part of a cross-sectional malaria study in Ghana, granzyme B levels and CD8⁺ T cells phenotypes were compared in the peripheral blood of children with complicated malaria, uncomplicated malaria, afebrile but asymptomatically infected children and non-infected children. Granzyme B levels in the plasma were significantly higher in children with febrile malaria than in afebrile children. CD8⁺ T cells were the main T cell subset expressing granzyme B. The proportion of granzyme B⁺ CD8⁺ T cells was significantly higher in children with complicated malaria than in uncomplicated malaria, whereas the activation marker CD38 on CD8⁺ T cells showed similar expression levels. This suggests a pathogenic role of cytotoxic CD8⁺ T cells in the development of malaria complications in humans.
... leukocytes themselves secrete proinflammatory cytokines, such as gamma interferon (IFN-␥), tumor necrosis factor (TNF), and interleukin 1 (IL-1), that further weaken the integrity of the blood-brain barrier (BBB) and exacerbate cerebral pathology (4)(5)(6)(7). ...
Article
Full-text available
We have used a genome-wide screen in N -ethyl- N -nitrosourea (ENU) mutagenized mice to identify genes in which recessive loss of function mutations protect against pathological neuroinflammation. We identified an R367Q mutation in the ZBTB7B (ThPOK) protein which homozygosity causes protection against experimental cerebral malaria (ECM) caused by infection with Plasmodium berghei ANKA (PbA). Zbtb7b R367Q homozygous mice show a defect in the lymphoid compartment expressed as severe reduction in the number of single positive CD4 T cells in the thymus and in the periphery, reduced brain infiltration of pro-inflammatory leukocytes in PbA infected mice and reduced production of pro-inflammatory cytokines by primary T cells ex vivo and in vivo . Dampening of proinflammatory immune responses in Zbtb7b R367Q mice is concomitant to increased susceptibility to infection with avirulent ( Mycobatcerium bovis , BCG) and virulent ( M. tuberculosis , H37Rv) mycobacteria. The R367Q mutation maps to the first DNA-binding zinc finger domain of ThPOK, and causes a loss of base contact by R367 in the major groove of DNA which is predicted to impair DNA binding. Global immunoprecipitation of ThPOK-containing chromatin complexes coupled to DNA sequencing (ChIP-seq) identified transcriptional networks and candidate genes likely to play a key role in CD4 ⁺ /CD8 ⁺ T cell development and in the expression of lineage specific functions of these cells. This study highlights ThPOK as a global regulator of immune function in which alterations may affect normal responses to infectious and inflammatory stimuli.
... A central hallmark of ECM is destruction of the blood-brain barrier (BBB) (12). It is now well-established that cytotoxic CD8 + T cells are the primary mediators of ECM development (13)(14)(15)(16)(17)(18)(19)(20)(21). During ECM, parasite-specific CD8 + T cells accumulate along cerebral vessels, where INF-γ release is thought to cause the activation of endothelial cells and perforin-mediated disruption of tight junctions to induce the BBB breakdown (20)(21)(22)(23)(24). ...
Article
Full-text available
Cerebral malaria is a life-threatening complication of malaria in humans, and the underlying pathogenic mechanisms are widely analyzed in a murine model of experimental cerebral malaria (ECM). Here, we show abrogation of ECM by hemocoel sporozoite-induced infection of a transgenic Plasmodium berghei line that overexpresses profilin, whereas these parasites remain fully virulent in transfusion-mediated blood infection. We, thus, demonstrate the importance of the clinically silent liver-stage infection for modulating the onset of ECM. Even though both parasites triggered comparable splenic immune cell expansion and accumulation of antigen-experienced CD8+ T cells in the brain, infection with transgenic sporozoites did not lead to cerebral vascular damages and suppressed the recruitment of overall lymphocyte populations. Strikingly, infection with the transgenic strain led to maintenance of CD115+Ly6C+ monocytes, which disappear in infected animals prone to ECM. An early induction of IL-10, IL-12p70, IL-6, and TNF at the time when parasites emerge from the liver might lead to a diminished induction of hepatic immunity. Collectively, our study reveals the essential role of early host interactions in the liver that may dampen the subsequent pro-inflammatory immune responses and influence the occurrence of ECM, highlighting a novel checkpoint in this fatal pathology.
... These results also suggest that experimental murine infections that mimic the CD8 + T cell-mediated cytolysis seen in patients may be better models for testing drugs than infection of C57BL/6 mice with Leishmania (7,8,21). In addition to CL, there are several infectious diseases where CD8 + cytolytic T cells have been implicated in promoting increased pathology-including cerebral malaria (39,40), trypanosomiasis (41), and coxsackievirus (42)raising the possibility that variability in the expression of genes associated with cytolysis might predict disease progression in these diseases as well. Furthermore, we have identified several genes that had not been previously known to influence leishmaniasis, and studies are ongoing to evaluate their role in disease. ...
Article
Patients infected with Leishmania braziliensis develop chronic lesions that often fail to respond to treatment with antiparasite drugs. To determine whether genes whose expression is highly variable in lesions between patients might influence disease outcome, we obtained biopsies of lesions from patients before treatment with pentavalent antimony and performed transcriptomic profiling on these clinical samples. We identified genes that were highly variably expressed between patients, and the variable expression of these genes correlated with treatment outcome. Among the most variable genes in all the patients were components of the cytolytic pathway, and the expression of these genes correlated with parasite load in the skin. We demonstrated that treatment failure was linked to the cytolytic pathway activated during infection. Using a host-pathogen marker profile of as few as three genes, we showed that eventual treatment outcome could be predicted before the start of treatment in two separate cohorts of patients with cutaneous leishmaniasis ( n = 21 and n = 25). These findings raise the possibility of point-of-care diagnostic screening to identify patients at high risk of treatment failure and provide a rationale for a precision medicine approach to drug selection in cutaneous leishmaniasis. This work more broadly demonstrates the value of identifying genes of high variability in other diseases to better understand and predict diverse clinical outcomes.
... A central hallmark of ECM is destruction of the blood-brain barrier (BBB) (12). It is now well-established that cytotoxic CD8 + T cells are the primary mediators of ECM development (13)(14)(15)(16)(17)(18)(19)(20)(21). During ECM, parasite-specific CD8 + T cells accumulate along cerebral vessels, where INF-γ release is thought to cause the activation of endothelial cells and perforin-mediated disruption of tight junctions to induce the BBB breakdown (20)(21)(22)(23)(24). ...
Article
Full-text available
Cerebral malaria is a life-threatening complication of malaria in humans, and the underlying pathogenic mechanisms are widely analyzed in a murine model of experimental cerebral malaria (ECM). Here, we show abrogation of ECM by hemocoel sporozoite-induced infection of a transgenic Plasmodium berghei line that overexpresses profilin, whereas these parasites remain fully virulent in transfusion-mediated blood infection. We, thus, demonstrate the importance of the clinically silent liver-stage infection for modulating the onset of ECM. Even though both parasites triggered comparable splenic immune cell expansion and accumulation of antigen-experienced CD8⁺ T cells in the brain, infection with transgenic sporozoites did not lead to cerebral vascular damages and suppressed the recruitment of overall lymphocyte populations. Strikingly, infection with the transgenic strain led to maintenance of CD115⁺Ly6C⁺ monocytes, which disappear in infected animals prone to ECM. An early induction of IL-10, IL-12p70, IL-6, and TNF at the time when parasites emerge from the liver might lead to a diminished induction of hepatic immunity. Collectively, our study reveals the essential role of early host interactions in the liver that may dampen the subsequent pro-inflammatory immune responses and influence the occurrence of ECM, highlighting a novel checkpoint in this fatal pathology.
... 367,368 Intravascular accumulation of platelets and immune cells has been observed in ECM, and CD8 + cells appear central to ECM pathogenesis. [369][370][371][372] The picture is less clear in CM, where some studies have reported infiltration of leukocytes and platelets within the brain microvasculature, 96,173 whereas others did not find that. 195,373 Intravascular accumulation of monocytes has also been reported, but there is little evidence of high numbers of CD8 + Tcell accumulation in human CM. ...
Article
Full-text available
Malaria, a mosquito‐borne infectious disease caused by parasites of the genus Plasmodium continues to be a major health problem worldwide. The unicellular Plasmodium‐parasites have the unique capacity to infect and replicate within host erythrocytes. By expressing variant surface antigens Plasmodium falciparum has evolved to avoid protective immune responses; as a result in endemic areas anti‐malaria immunity develops gradually over many years of multiple and repeated infections. We are studying the role of Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) expressed by asexual stages of P. falciparum responsible for the pathogenicity of severe malaria. The immunopathology of falciparum malaria has been linked to cyto‐adhesion of infected erythrocytes to specific host receptors. A greater appreciation of the PfEMP1 molecules important for the development of protective immunity and immunopathology is a prerequisite for the rational discovery and development of a safe and protective anti‐disease malaria vaccine. Here we review the role of ICAM‐1 and EPCR receptor adhering falciparum‐parasites in the development of severe malaria; we discuss our current research to understand the factors involved in the pathogenesis of cerebral malaria and the feasibility of developing a vaccine targeted specifically to prevent this disease.
... The ECM mouse model replicates the neurological symptoms, parasite accumulation, and leukocyte infiltration observed in children with CM, including the induction of brain oedema and brain stem death as a predictor of severe disease [75][76][77]. Numerous studies in both mice and humans have demonstrated that an excessive pro-inflammatory response results in severe immunopathology during malaria [78][79][80]. IFNγwhich can be secreted in large amounts by NK cells-is a critical mediator of immune-mediated damage during ECM [81,82]. An initial report using anti-NK1.1 ...
Article
Full-text available
Natural killer (NK) cells are important innate effector cells that are well described in their ability to kill virally-infected cells and tumors. However, there is increasing appreciation for the role of NK cells in the control of other pathogens, including intracellular parasites such as Plasmodium, the cause of malaria. NK cells may be beneficial during the early phase of Plasmodium infection-prior to the activation and expansion of antigen-specific T cells-through cooperation with myeloid cells to produce inflammatory cytokines like IFNγ. Recent work has defined how Plasmodium can activate NK cells to respond with natural cytotoxicity, and inhibit the growth of parasites via antibody-dependent cellular cytotoxicity mechanisms (ADCC). A specialized subset of adaptive NK cells that are negative for the Fc receptor γ chain have enhanced ADCC function and correlate with protection from malaria. Additionally, production of the regulatory cytokine IL-10 by NK cells prevents overt pathology and death during experimental cerebral malaria. Now that conditional NK cell mouse models have been developed, previous studies need to be reevaluated in the context of what is now known about other immune populations with similarity to NK cells (i.e., NKT cells and type I innate lymphoid cells). This brief review summarizes recent findings which support the potentially beneficial roles of NK cells during Plasmodium infection in mice and humans. Also highlighted are how the actions of NK cells can be explored using new experimental strategies, and the potential to harness NK cell function in vaccination regimens.
Article
Gout is characterized by hyperuricemia and recurrent inflammatory episodes caused by intra-articular crystal deposition of monosodium urate (MSU). There is a clear relationship between gout and metabolic syndrome. Recent evidence indicates that perforin plays a role in regulating glucose homeostasis and provides protection in diet-induced non-alcoholic steatohepatitis models. However, the impact of perforin on immune inflammation in gout remains unclear. We induced acute gout models in both wild-type (WT) mice and Prf1null mice by administering intra-articular injections of MSU crystals. We compared the ankle joint swelling and the histological score between the two groups. Furthermore, we investigated underlying mechanisms through in vitro co-culture experiments involving CD8 T cells and macrophages. In this study, Prf1null mice showed significantly more pronounced ankle swelling with increased inflammatory cell infiltrations compared with WT mice 24 h after local MSU injection. Moreover, MSU-induced Prf1null mice exhibited increased accumulation of CD8 T cells but not NK cells. Perforin-deficient CD8 T cells displayed reduced cytotoxicity towards bone marrow–derived M0 and M1 macrophages and promoted TNF-α secretion from macrophage. Perforin from CD8 T cells limits joint inflammation in mice with acute gout by downregulating macrophage-mediated inflammation. Key Points • Perforin deficiency increased swelling in the ankle joints of mice upon MSU injection. • Perforin deficiency is associated with increased immune cell recruitment and severe joint damage in gout. • Perforin regulated CD8 T cell accumulation in gout and promoted CD8 T cell cytotoxicity towards M0 and M1 macrophages. • CD8 T cell-derived perforin regulated pro-inflammatory cytokine secretion of macrophage.
Article
Full-text available
The Cytokine-inducible Src homology 2 domain-containing (CISH) protein is a negative feedback regulator induced by cytokines that play key roles in immunity and erythropoiesis. Single nucleotide polymorphisms (SNPs) in the human CISH gene have been associated with increased susceptibility to severe malaria disease. To directly assess how CISH might influence outcomes in the BALB/c model of malaria anemia, CISH knockout (Cish−/−) mice on this background were infected with Plasmodium berghei and their hematopoietic responses, cytokine production and ability to succumb to severe malaria disease evaluated. Despite basal erythrocytic disruption, upon P. berghei infection, the Cish −/− mice were better able to maintain peripheral blood cell counts, hemoglobin levels and a steady-state pattern of erythroid differentiation compared to wild-type (Cish+/+) mice. Ablation of CISH, however, did not influence the outcome of acute malaria infections in either the BALB/c model or the alternative C57BL/6 model of experimental cerebral malaria, with the kinetics of infection, parasite load, weight loss and cytokine responses being similar between Cish+/+ and Cish−/− mice, and both genotypes succumbed to experimental cerebral malaria within a comparable timeframe.
Article
Full-text available
Introduction It is well known that dietary changes have a significant impact on the immune system, and modifications in lipid balance may contribute to disease progression in several cases. Malaria is still a major global health concern, and the development of the disease has already been linked to the host’s nutritional status, so it’s critical to understand how environmental factors, such as dietary variations, can influence the outcome of infection. We therefore investigated the effect of a short-term diet in a murine model of experimental cerebral malaria. Methods For this, male C57BL/6 mice were fed a high fat diet containing 60% of the calories from lipids for 5 days. Following this period, the animals were infected with Plasmodium berghei ANKA, and parasitemia, survival, and neurological scores were compared. Considering that one of the first elimination routes of the intracellular parasite is oxidative stress, the antioxidant N-acetylcysteine was administered to assess whether the protection would be reversed. Results and discussion Animals fed a hyperlipidic diet reacted the same way to infection even after NAC administration. Unlike the control group, which died after eight days of infection with roughly 7% parasitized red blood cells, the hyperlipidic diet group was resistant to infection, with no clinical signs and no increase in blood parasitemia. Several proinflammatory cytokines such as TNF-α IFN-γ and IL-6 were increased in the spleen of both infected groups, regardless of their diet. The provision of a high-fat diet to mice for as little as 5 days completely prevents Plasmodium berghei ANKA infection in C57BL/6 mice, while the treatment of an antioxidant failed to reverse the parasite protection.
Article
Malaria, the most prevalent mosquito-borne infectious disease worldwide, has accompanied humanity for millennia and remains an important public health issue despite advances in its prevention and treatment. Most infections are asymptomatic, but a small percentage of individuals with a heavy parasite burden develop severe malaria, a group of clinical syndromes attributable to organ dysfunction. Cerebral malaria is an infrequent but life-threatening complication of severe malaria that presents as an acute cerebrovascular encephalopathy characterized by unarousable coma. Despite effective antiparasite drug treatment, 20% of patients with cerebral malaria die from this disease, and many survivors of cerebral malaria have neurocognitive impairment. Thus, an important unmet clinical need is to rapidly identify people with malaria who are at risk of developing cerebral malaria and to develop preventive, adjunctive and neuroprotective treatments for cerebral malaria. This Review describes important advances in the understanding of cerebral malaria over the past two decades and discusses how these mechanistic insights could be translated into new therapies.
Article
Cerebral Malaria (CM) is associated with the complex neurological syndrome, whose pathology is mediated by severe inflammatory processes following infection with Plasmodium falciparum. Coenzyme-Q10 (Co-Q10) is a potent anti-inflammatory, anti-oxidant, and anti-apoptotic agent with numerous clinical applications. The aim of this study was to elucidate the role of oral administration of Co-Q10 on the initiation or regulation of inflammatory immune response during experimental cerebral malaria (ECM). For this purpose, the pre-clinical effect of Co-Q10 was evaluated in C57BL/6J mice infected with Plasmodium berghei ANKA (PbA). Treatment with Co-Q10 resulted in the reduction of infiltrating parasite load, greatly improved the survival rate of PbA-infected mice that occurred independent of parasitaemia and prevented PbA-induced disruption of the blood-brain barrier (BBB) integrity. Exposure to Co-Q10 resulted in the reduction of infiltration of effector CD8+ T cells in the brain and secretion of cytolytic Granzyme B molecules. Notably, Co-Q10-treated mice had reduced levels of CD8+T cell chemokines CXCR3, CCR2, and CCR5 in the brain following PbA-infection. Brain tissue analysis showed a reduction in the levels of inflammatory mediators TNF- α, CCL3, and RANTES in Co-Q10 administered mice. In addition, Co-Q10 modulated the differentiation and maturation of both splenic and brain dendritic cells and cross-presentation (CD8α+DCs) during ECM. Remarkably, Co-Q10 was very effective in decreasing levels of CD86, MHC-II, and CD40 in macrophages associated with ECM pathology. Exposure to Co-Q10 resulted in increased expression levels of Arginase-1 and Ym1/chitinase 3-like 3, which is linked to ECM protection. Furthermore, Co-Q10 supplementation prevented PbA-induced depletion of Arginase and CD206 mannose receptor levels. Co-Q10 abrogated PbA-driven elevation in pro-inflammatory cytokines IL-1β, IL-18, and IL-6 levels. In conclusion, the oral supplementation with Co-Q10 decelerates the occurrence of ECM by preventing lethal inflammatory immune responses and dampening genes associated with inflammation and immune-pathology during ECM, and offers an inimitable opening for developing an anti-inflammatory agent against cerebral malaria.
Article
The immune system performs critical functions to defend against invading pathogens and maintain tissue homeostasis. Immune cells reside within or are recruited to a host of mechanically active tissues throughout the body and, as a result, are exposed to varying types and degrees of mechanical stimuli. Despite their abundance in such tissues, the role of mechanical stimuli in influencing immune cell function and the molecular mechanisms responsible for mechanics-mediated changes are still poorly understood. The recent emergence of mechanically-gated ion channels, particularly Piezo1, has provided an exciting avenue of research within the fields of mechanobiology and immunology. Numerous studies have identified roles for mechanically-gated ion channels in mechanotransduction within various different cell types, with a few recent studies in immune cells. These initial studies provide strong evidence that mechanically-gated ion channels play pivotal roles in regulating the immune system. In this review, we discuss characteristics of ion channel mediated mechanotransduction, review the current techniques used to quantify and visualize ion channel activity in response to mechanical stimuli, and finally we provide an overview of recent studies examining the role of mechanically-gated ion channels in modulating immune cell function.
Article
In the Plasmodium berghei ANKA mouse model of malaria, accumulation of CD8+ T cells and infected RBCs in the brain promotes the development of experimental cerebral malaria (ECM). In this study, we used malaria-specific transgenic CD4+ and CD8+ T cells to track evolution of T cell immunity during the acute and memory phases of P. berghei ANKA infection. Using a combination of techniques, including intravital multiphoton and confocal microscopy and flow cytometric analysis, we showed that, shortly before onset of ECM, both CD4+ and CD8+ T cell populations exit the spleen and begin infiltrating the brain blood vessels. Although dominated by CD8+ T cells, a proportion of both T cell subsets enter the brain parenchyma, where they are largely associated with blood vessels. Intravital imaging shows these cells moving freely within the brain parenchyma. Near the onset of ECM, leakage of RBCs into areas of the brain can be seen, implicating severe damage. If mice are cured before ECM onset, brain infiltration by T cells still occurs, but ECM is prevented, allowing development of long-term resident memory T cell populations within the brain. This study shows that infiltration of malaria-specific T cells into the brain parenchyma is associated with cerebral immunopathology and the formation of brain-resident memory T cells. The consequences of these resident memory populations is unclear but raises concerns about pathology upon secondary infection.
Article
Full-text available
Thorough understanding of the role of CD4 T cells in immunity can be greatly assisted by the study of responses to defined specificities. This requires knowledge of Plasmodium-derived immunogenic epitopes, of which only a few have been identified, especially for the mouse C57BL/6 background. We recently developed a TCR transgenic mouse line, termed PbT-II, that produces CD4⁺ T cells specific for an MHC class II (I-Ab)-restricted Plasmodium epitope and is responsive to both sporozoites and blood-stage P. berghei. Here, we identify a peptide within the P. berghei heat shock protein 90 as the cognate epitope recognised by PbT-II cells. We show that C57BL/6 mice infected with P. berghei blood-stage induce an endogenous CD4 T cell response specific for this epitope, indicating cells of similar specificity to PbT-II cells are present in the naïve repertoire. Adoptive transfer of in vitro activated TH1-, or particularly TH2-polarised PbT-II cells improved control of P. berghei parasitemia in C57BL/6 mice and drastically reduced the onset of experimental cerebral malaria. Our results identify a versatile, potentially protective MHC-II restricted epitope useful for exploration of CD4 T cell-mediated immunity and vaccination strategies against malaria.
Article
Cerebral malaria (CM) is one of the most severe pathologies of malaria; it induces neuro-cognitive sequelae and has a high mortality rate. Although many factors involved in the development of CM have been discovered, its pathogenic mechanisms are still not completely understood. Most studies on CM have focused on the blood–brain barrier (BBB), despite the importance of the blood–cerebrospinal fluid barrier (BCSFB), which protects the brain from peripheral inflammation. Consequently, the pathological role of the BCSFB in CM is currently unknown. To examine the status of the BCSFB in CM and malaria without this pathology (non-CM), we developed a new method for evaluating the permeabilization of the BCSFB during CM in mice, using Evans blue dye and a software-assisted image analysis. Using C57BL/6J (B6) mice infected with Plasmodium berghei ANKA strain as an experimental CM model and B6 mice infected with P. berghei NK65 strain or Plasmodium yoelii as non-CM models, we revealed that the permeability of the BCSFB increased during experimental CM but not during non-CM. We observed hemorrhaging in the cerebral ventricles and hemozoin-like structures in the choroid plexus, which is a key component of the BCSFB, in CM mice. Taken together, this evidence indicates that the BCSFB is disrupted in experimental CM, whereas it remains intact in non-CM. We also found that P. berghei ANKA parasites and CD8⁺ T cells are involved in the BCSFB disruption in experimental CM. An understanding of the mechanisms underlying CM might help in the development of effective strategies to prevent and manage CM in humans.
Article
Background Cerebral malaria (CM) is the most severe complication in malaria. Endothelial activation, cytokine release and vascular obstruction are essential hallmarks of CM. Clinical studies have suggested a link between von Willebrand factor (VWF) and malaria pathology. Objectives To investigate the contribution of VWF in the pathogenesis of experimental cerebral malaria (ECM). Methods Both Vwf+/+ and Vwf‐/‐ mice were infected with Plasmodium berghei ANKA (Pb ANKA) to induce ECM. Alterations of plasma VWF and ADAMTS13, platelet count, neurological features and accumulation of platelets and leukocytes in the brain were examined following infection. Results Plasma VWF levels significantly increased upon Pb ANKA infection in Vwf+/+ animals. While ADAMTS13 activity was not affected, high molecular weight VWF multimers disappeared at the end‐stage ECM, possibly due to an ongoing hypercoagulability. Although the number of reticulocytes, a preferential target for the parasites, was increased in Vwf‐/‐ mice compared to Vwf+/+ mice early after infection, parasitemia levels did not markedly differ between the 2 groups. Interestingly, Vwf‐/‐ mice manifested overall clinical ECM features similar to those observed in Vwf+/+ animals. At day 8.5 post‐infection, however, clinical ECM features in Vwf‐/‐ mice were slightly more beneficial than in Vwf+/+ animals. Despite these minor differences, overall survival was not different between Vwf‐/‐ and Vwf+/+ mice. Similarly, Pb ANKA‐induced thrombocytopenia, leukocyte and platelet accumulations in the brains, were not altered by the absence of VWF. Conclusions Our study suggests that increased VWF concentration is a hallmark of ECM. However, VWF does not have a major influence in modulating late‐stage ECM pathogenesis.
Article
Full-text available
Two molecular mechanisms of T cell-mediated cytotoxicity, one perforin-based, the other Fas-based, have been demonstrated. To determine the extent of their contribution to T cell-mediated cytotoxicity, a range of effector cells from normal control or perforin-deficient mice were tested against a panel of target cells with various levels of Fas expression. All cytotoxicity observed was due to either of these mechanisms, and no third mechanism was detected. Thus, the perforin- and Fas-based mechanisms may account for all T cell-mediated cytotoxicity in short-term in vitro assays.
Article
Full-text available
Fas ligand (FasL), a cell surface molecule belonging to the tumor necrosis factor family, binds to its receptor Fas, thus inducing apoptosis of Fas-bearing cells. Various cells express Fas, whereas FasL is expressed predominantly in activated T cells. In the immune system, Fas and FasL are involved in down-regulation of immune reactions as well as in T cell-mediated cytotoxicity. Malfunction of the Fas system causes lymphoproliferative disorders and accelerates autoimmune diseases, whereas its exacerbation may cause tissue destruction.
Article
Full-text available
Chemokines direct tissue invasion by specific leukocyte populations. Thus, chemokines may play a role in multiple sclerosis (MS), an idiopathic disorder in which the central nervous system (CNS) inflammatory reaction is largely restricted to mononuclear phagocytes and T cells. We asked whether specific chemokines were expressed in the CNS during acute demyelinating events by analyzing cerebrospinal fluid (CSF), whose composition reflects the CNS extracellular space. During MS attacks, we found elevated CSF levels of three chemokines that act toward T cells and mononuclear phagocytes: interferon-gamma-inducible protein of 10 kDa (IP-10); monokine induced by interferon-gamma (Mig); and regulated on activation, normal T-cell expressed and secreted (RANTES). We then investigated whether specific chemokine receptors were expressed by infiltrating cells in demyelinating MS brain lesions and in CSF. CXCR3, an IP-10/Mig receptor, was expressed on lymphocytic cells in virtually every perivascular inflammatory infiltrate in active MS lesions. CCR5, a RANTES receptor, was detected on lymphocytic cells, macrophages, and microglia in actively demyelinating MS brain lesions. Compared with circulating T cells, CSF T cells were significantly enriched for cells expressing CXCR3 or CCR5. Our results imply pathogenic roles for specific chemokine-chemokine receptor interactions in MS and suggest new molecular targets for therapeutic intervention.
Article
Full-text available
Multiple sclerosis (MS) is a T cell-dependent chronic inflammatory disease of the central nervous system. The role of chemokines in MS and its different stages is uncertain. Recent data suggest a bias in expression of chemokine receptors by Th1 vs. Th2 cells; human Th1 clones express CXCR3 and CCR5 and Th2 clones express CCR3 and CCR4. Chemokine receptors expressed by Th1 cells may be important in MS, as increased interferon-gamma (IFN-gamma) precedes clinical attacks, and IFN-gamma injection induces disease exacerbations. We found CXCR3(+) T cells increased in blood of relapsing-remitting MS, and both CCR5(+) and CXCR3(+) T cells increased in progressive MS compared with controls. Furthermore, peripheral blood CCR5(+) T cells secreted high levels of IFN-gamma. In the brain, the CCR5 ligand, MIP-1alpha, was strongly associated with microglia/macrophages, and the CXCR3 ligand, IP-10, was expressed by astrocytes in MS lesions but not unaffected white matter of control or MS subjects. Areas of plaque formation were infiltrated by CCR5-expressing and, to a lesser extent, CXCR3-expressing cells; Interleukin (IL)-18 and IFN-gamma were expressed in demyelinating lesions. No leukocyte expression of CCR3, CCR4, or six other chemokines, or anti-inflammatory cytokines IL-5, IL-10, IL-13, and transforming growth factor-beta was observed. Thus, chemokine receptor expression may be used for immunologic staging of MS and potentially for other chronic autoimmune/inflammatory processes such as rheumatoid arthritis, autoimmune diabetes, or chronic transplant rejection. Furthermore, these results provide a rationale for the use of agents that block CCR5 and/or CXCR3 as a therapeutic approach in the treatment of MS.
Article
Full-text available
To characterize the T cells involved in the pathogenesis of cerebral malaria (CM) induced by infection with Plasmodium berghei ANKA clone 1.49L (PbA 1.49L), the occurrence of the disease was assessed in mice lacking T cells of either the alphabeta or gammadelta lineage (TCRalphabeta(-/-) or TCRgammadelta(-/-)). TCRgammadelta(-/-) mice were susceptible to CM, whereas all TCRalphabeta(-/-) mice were resistant, suggesting that T cells of the alphabeta lineage are important in the genesis of CM. The repertoire of TCR V(beta) segment gene expression was examined by flow cytometry in B10.D2 mice, a strain highly susceptible to CM induced by infection with PbA 1.49L. In these mice, CM was associated with an increase of T cells bearing the V(beta)8.1, 2 segments in the peripheral blood lymphocytes. Most V(beta)8.1, 2(+) T cells from peripheral blood lymphocytes of the mice that developed CM belonged to the CD8 subset, and exhibited the CD69(+), CD44(high) and CD62L(low) phenotype surface markers. The link between the increase in V(beta)8.1, 2(+) T cells and the neuropathological consequences of PbA infection was strengthened by the observation that the occurrence of CM was significantly reduced in mice treated with KJ16 antibodies against the V(beta)8.1 and V(beta)8.2 chains, and in mice rendered deficient in V(beta)8.1(+) T cells by a mouse mammary tumor virus superantigen.
Article
Full-text available
Malaria still is a major public health problem, partly because the pathogenesis of its major complication, cerebral malaria, remains incompletely understood. Experimental models represent useful tools to better understand the mechanisms of this syndrome. Here, data generated by several models are reviewed both in vivo and in vitro; we propose that some pathogenic mechanisms, drawn from data obtained from experiments in a mouse model, may be instrumental in humans. In particular, tumor necrosis factor (TNF) receptor 2 is involved in this syndrome, implying that the transmembrane form of TNF may be more important than the soluble form of the cytokine. It has also been shown that in addition to differences in immune responsiveness between genetically resistant and susceptible mice, there are marked differences at the level of the target cell of the lesion, namely, the brain endothelial cell. In murine cerebral malaria, a paradoxical role of platelets has been proposed. Indeed, platelets appear to be pathogenic rather than protective in inflammatory conditions because they can potentiate the deleterious effects of TNF. More recently, it has been shown that interactions among platelets, leukocytes, and endothelial cells have phenotypic and functional consequences for the endothelial cells. A better understanding of these complex interactions leading to vascular injury will help improve the outcome of cerebral malaria.
Article
Full-text available
The CXC chemokine ligand (CXCL)10 is induced locally in the CNS in diverse pathologic states. The impact of CXCL10 production in the CNS was examined in transgenic mice with astrocyte-directed production of this chemokine. These glial fibrillary acidic protein (GF)-CXCL10 transgenic mice spontaneously developed transgene dose- and age-related leukocyte infiltrates in perivascular, meningeal, and ventricular regions of the brain that were composed of, surprisingly, mainly neutrophils and, to a lesser extent, T cells. No other overt pathologic or physical changes were evident. In addition, the cerebral expression of a number of inflammation-related genes (e.g., cytokines) was not significantly altered in the transgenic mice. The extent of leukocyte recruitment to the brain could be enhanced markedly by peripheral immunization of GF-CXCL10 mice with CFA and pertussis toxin. This was paralleled by a modest, transient increase in the expression of some cytokine and chemokine genes. Analysis of the expression of the CXCL10 receptor, CXCR3, by the brain-infiltrating leukocytes from immunized GF-CXCL10 transgenic mice revealed a significant enrichment for CXCR3-positive cells in the CNS compared with spleen. The majority of cells positive for CXCR3 coexpressed CD3, whereas Gr1-positive granulocytes were negative for CXCR3 expression. Thus, while astrocyte production of CXCL10 can promote spontaneous and potentiate immune-induced recruitment of leukocytes to the CNS, this is not associated with activation of a degenerative immune pathology. Finally, the accumulation of neutrophils in the brain of GF-CXCL10 transgenic mice is apparently independent of CXCR3 and involves an unknown mechanism.
Article
lated in EAE, were undetectable in IFN-g- and IFN-gR-deficient mice. Macrophage inflammatory protein-2 and T cell activation gene-3, both neutrophil-attracting chemokines, were strongly up-regulated. There was no induction of the Th2 cytokines, IL-4, IL-10, or IL-13. RNase protection assays and RT-PCR showed the prevalence of IL-2, IL-3, and IL-15, but no increase in IL-12p40 mRNA levels in IFN-g- or IFN-gR-deficient mice with EAE. Lymph node cells from IFN-g-deficient mice proliferated in response to myelin basic protein, whereas BALB/c lymph node cells did not. These findings show a regulatory role for IFN-g in EAE, acting on T cell proliferation and directing chemokine production, with profound implications for the onset and progression of disease. The Journal of Immunology, 2000, 164: 2759 -2768.
Article
IFN- has been implicated in the pathogenesis of experimental cerebral malaria (ECM). We have used mice lacking the chain of the IFN- receptor (KO mice) to define its role in the pathogenesis of ECM. Infected KO mice did not develop ECM and showed no leukocyte or parasite sequestration in the brain, and no hemorrhages. The resistance of KO mice to ECM was associated with the absence of any increases of TNF- and ICAM-1 proteins in the brain, which are both essential for ECM. Wild-type (WT) mice which do not develop ECM, despite increased local production of TNF- protein, showed no leukocyte accumulation in the brain and this was correlated with the absence of ICAM-1 protein from brain microvessels. KO mice infected with 106 parasitized erythrocytes (PE) of Plasmodium berghei ANKA (PbA) did not develop ECM, but they had high parasitemia and died earlier than WT mice which did not develop ECM. However, KO mice did not develop higher parasitemia than WT mice when both groups were infected with a lower dose (5×105 PE) of PbA-infected red blood cells. This indicates that different doses of PE may trigger different IFN- responses and that there may be a threshold concentration for protection against parasitemia.
Article
In vitro, T cell-dependent cytotoxicity is mediated by two distinct mechanisms, one being perforin-, the other Fas-dependent. The contribution of both of these mechanisms to clearance of viral infections was investigated in mice for the noncytopathic lymphocytic choriomeningitis virus (LCMV) and the cytopathic vaccinia, vesicular stomatitis (VSV) and Semliki forest (SFV) viruses. Clearance of an acute LCMV infection was mediated by the perforin-dependent mechanism without measurable involvement of the Fas-dependent pathway. For the resolution of vaccinia virus infection and for resistance against VSV and SFV, however, neither of the two pathways was required. These data suggest that perforin-dependent cytotoxicity mediated by T cells is crucial for protection against noncytopathic viruses, whereas infections with cytopathic viruses are controlled by nonlytic T cell-dependent soluble mediators such as cytokines (IFN-γ against vaccinia virus) and neutralizing antibodies (against VSV and SFV).
Article
Macrophage inflammatory protein (MIP)-1α is a chemokine that is associated with Th1 cytokine responses. Expression and antibody blocking studies have implicated MIP-1α in multiple sclerosis (MS) and in experimental autoimmune encephalomyelitis (EAE). We examined the role of MIP-1α and its CCR5 receptor in the induction of EAE by immunizing C57BL / 6 mice deficient in either MIP-1α or CCR5 with myelin oligodendrocyte glycoprotein (MOG). We found that MIP-1α-deficient mice were fully susceptible to MOG-induced EAE. These knockout animals were indistinguishable from wild-type mice in Th1 cytokine gene expression, the kinetics and severity of disease, and infiltration of the central nervous system by lymphocytes, macrophages and granulocytes. RNase protection assays showed comparable accumulation of mRNA for the chemokines interferon-inducible protein-10, RANTES, macrophage chemoattractant protein-1, MIP-1β, MIP-2, lymphotactin and T cell activation gene-3 during the course of the disease. CCR5-deficient mice were also susceptible to disease induction by MOG. The dispensability of MIP-1α and CCR5 for MOG-induced EAE in C57BL / 6 mice supports the idea that differential chemokine expression patterns represent differences in disease mechanism that underlie various models of EAE, and possibly distinct patterns of pathology seen in MS.
Article
In susceptible mouse strains, infection of mice with Plasmodium berghei ANKA (PbA) results in a lethal complication, cerebral malaria. Cerebral malaria is due to the immune response induced by the parasite, which results in an increased production of TNF, known to increase the expression of adhesion molecules on the endothelia. To investigate the role of the adhesion molecule ICAM-1 (CD54), we infected wild-type (+/+) and ICAM-1-deficient (-/-) mice with PbA. While +/+ mice died 6–8 days after infection, -/- mice survived > 15 days. Parasitaemia was similar in +/+ and -/- mice. Serum TNF concentration was increased by the infection and was significantly higher in infected +/+ than in -/- mice. However, TNF mRNA levels in spleen, lungs, and brain were elevated in both infected +/+ and -/- mice. For IFN-γ, serum levels were similar in both groups. A breakdown of the blood-brain barrier was evident in infected +/+ mice only. Interestingly, thrombocytopenia was profound in infected +/+, but practically absent in -/- mice. Moreover, macrophage sequestration was evident in brain venules and lung capillaries of +/+ mice and was significantly less important in the alveolar capillaries of infected -/- mice. In contrast, neutrophil sequestration in the lung was similar in both +/+ and -/- mice. Sequestration of parasitized red blood cells was significantly greater in the alveolar capillaries from +/+ than -/- mice. These results indicate that while the immune response is similar in both +/+ and ICAM-1-/- mice, the absence of mortality in ICAM-/- mice correlates with a decrease of macrophage and parasitized RBC trapping and a less severe thrombocytopenia.
Article
Although the mechanisms of human immunodeficiency virus (HIV) neuroinvasion, neuronal injury, and subsequent development of HIV-1-associated AIDS dementia complex are not fully understood, a correlation between monocyte/macrophage infiltrates in the brain and neurological disease exists. In light of the many potential roles that chemokines and chemokine receptors may play in HIV neuropathogenesis, we sought to describe their pattern of expression in the SIV-infected rhesus macaque model of HIV encephalitis. We previously demonstrated elevated expression of the chemokines macrophage inflammatory protein (MIP)-1alpha, MIP-1beta, RANTES, and interferon-inducible protein (IP)-10 in brain of macaque monkeys with SIV encephalitis. In this study, we demonstrate that the corresponding chemokine receptors CCR3, CCR5, CXCR3, and CXCR4 are expressed in perivascular infiltrates in these same tissues. In addition, we detected CCR3, CCR5, and CXCR4 on subpopulations of large hippocampal and neocortical pyramidal neurons and on glial cells in both normal and encephalitic brain. These findings suggest that multiple chemokines and their receptors contribute to monocyte and lymphocyte recruitment to the brain in SIV encephalitis. Furthermore, the expression of known HIV/SIV co-receptors on neurons suggests a possible mechanism whereby HIV or SIV can directly interact with these cells, disrupting their normal physiological function and contributing to the pathogenesis of AIDS dementia complex.
Article
Blockage of the cerebral microvasculature by Plasmodium falciparum-infected erythrocytes appears to be the principal cause of human cerebral malaria. Knobs which appear on the membrane of the infected erythrocytes adhere to the endothelium, causing the obstruction of cerebral microvessels. Protein molecules such as CD36, thrombospondin, and intercellular adhesion molecule-1, which are present on the membrane of endothelial cells, may act as receptors for the attachment of knobs of P. falciparum-infected erythrocytes. Each of these candidate host molecules for infected-cell recognition and attachment are expressed in microvessels of the human brain. The presence of HRP1 and HRP2 in the cerebral microvessels of cerebral malaria patients may indicate the involvement of knob proteins in the pathogenesis of cerebral malaria. Owl monkeys infected with P. falciparum do not develop cerebral malaria. There is no blockage of cerebral microvessels by infected erythrocytes and knob proteins are absent. These findings support the contention that cerebral microvessel blockage and the presence of knob proteins are the probable causes of cerebral malaria.
Article
CTL specific for HIV have been described in lungs of infected patients at early stages of HIV disease. In order to characterize the evolution over time of HIV-specific CTL, we have analyzed the cytotoxic function and the cell surface phenotype of the alveolar lymphocytes from 41 patients at various stages of HIV disease. We demonstrated a progressive decline of alveolar anti-HIV CTL activity and detected Ts cells from the lungs of patients with advanced HIV disease. These alveolar T cells strongly suppressed the effector phase of anti-HIV CTL lysis. They lacked a marked specificity of function because they also block anti-HLA CTL response and were not restricted by the HLA-class-I transplantation Ag. They displayed the CD3, CD8, and HNK1 markers, were CD4 and CD16 negative, and lacked NK activity. The presence of Ts cells at late stages of HIV disease could thus partly explain the inefficiency of host defenses against HIV.
Article
The course of lethal Plasmodium berghei infection was examined in nu/+ and T cell-deficient nu/nu BALB/c mice. A rapidly fatal neurologic syndrome, including ataxia, hemiparesis, and seizures, was seen in the nu/+ mice early in the infection, whereas this syndrome was absent in the nu/nu mice. The nu/nu mice also developed anemia more slowly, had lower levels of immune complexes and total IgG, and had smaller decreases in serum C3 compared with the nu/+ mice. Histopathologic examination of the brains revealed cerebral malaria lesions, including vascular plugging and micro-hemorrhages, in the nu/+ mice but not in the nu/nu mice. Cerebral lesions similar in frequency and severity to those in nu/+ mice developed in nu/nu mice given spleen cells from normal nu/+ mice. The results suggest that an intact immune system is necessary for the expression of cerebral malaria.
Article
Guidelines for submitting commentsPolicy: Comments that contribute to the discussion of the article will be posted within approximately three business days. We do not accept anonymous comments. Please include your email address; the address will not be displayed in the posted comment. Cell Press Editors will screen the comments to ensure that they are relevant and appropriate but comments will not be edited. The ultimate decision on publication of an online comment is at the Editors' discretion. Formatting: Please include a title for the comment and your affiliation. Note that symbols (e.g. Greek letters) may not transmit properly in this form due to potential software compatibility issues. Please spell out the words in place of the symbols (e.g. replace “α” with “alpha”). Comments should be no more than 8,000 characters (including spaces ) in length. References may be included when necessary but should be kept to a minimum. Be careful if copying and pasting from a Word document. Smart quotes can cause problems in the form. If you experience difficulties, please convert to a plain text file and then copy and paste into the form.
Article
The recent generation of perforin knock-out mice has demonstrated a crucial role for the pore-forming perforin in cytolytic T-lymphocyte (CTL)-mediated cytolysis. Perforin-deficient mice failed to clear lymphocytic choriomeningitis virus in vivo, yet substantial killing activity still remained in perforin-free CTLs in vitro, indicating the presence of (a) further lytic pathway(s). Fas is an apoptosis-signalling receptor molecule on the surface of a number of different cells. Here we report that both perforin-deficient and Fas-ligand-deficient CTLs show impaired lytic activity on all target cells tested. The killing activity was completely abolished when both pathways were inactivated by using target cells from Fas-receptor-deficient lpr mice and perforin-free CTL effector cells. Fas-ligand-based killing activity was triggered upon T-cell receptor occupancy and was directed to the cognate target cell. Thus, two complementary, specific cytotoxic mechanisms are functional in CTLs, one based on the secretion of lytic proteins and one which depends on cell-surface ligand-receptor interaction.
Article
Cell surface molecules have received intense attention in recent years because of the central roles they play at the interface between the external environment and the cellular interior. Their functions include adhesion to other cells or extracellular matrices, protection against hostile physical, chemical and biological agents and the transport of metabolites into and out of the cell. In addition, cell surface molecules transduce signals across the cell membrane, relaying information inwards and presenting altered characteristics to the exterior as the environment changes.
Article
Studies with perforin-deficient mice have demonstrated that two independent mechanisms account for T cell-mediated cytotoxicity: A main pathway is mediated by the secretion of the pore-forming protein perforin by the cytotoxic T cell, whereas an alternative nonsecretory pathway relies on the interaction of the Fas ligand that is upregulated during T cell activation with the apoptosis-inducing Fas molecule on the target cell. NK cells use the former pathway exclusively. The protective role of the perforin-dependent pathway has been shown for infection with the noncytopathic lymphocytic choriomeningitis virus, for infection with Listeria monocytogenes, and for the elimination of tumor cells by T cells and NK cells. In contrast, perforin-dependent cytotoxicity is not involved in protection against the cytopathic vaccinia virus and vesicular stomatitis virus. LCMV-induced immunopathology and autoimmune diabetes have been found to require perforin-expression. A contribution of perforin-dependent cytotoxicity to the rejection of MHC class I-disparate heart grafts has also been observed. Its absence is efficiently compensated in rejection of fully allogeneic organ or skin grafts. So far, evidence for a role of Fas-dependent cytotoxicity as a T cell effector mechanism in vivo is lacking. Current data suggest that the main function of Fas may be in regulation of the immune response and apparently less at the level of an effector mechanism in host defense. Further analysis is necessary, however, to settle this point finally.
Article
We determined the requirement for selected lymphocyte subsets and cytokines in the pathogenesis of experimental murine cerebral malaria (CM) by using gene-targeted knockout and mAb-suppressed mice. Plasmodium berghei ANKA infection induced CM in A 0/0 mice, which lack expression of surface MHC class II glycoproteins and consequently express a severe and chronic reduction in numbers of CD4+ T cells. However, when A 0/0 mice, which are on a C57BL/6 x 129 genetic background, or immune-intact C57BL/6 controls treated with anti-CD4 mAb were infected, none developed CM. The latter finding confirms an earlier report that CD4+ T cells are required for CM to occur and additionally indicates that the reduced numbers of CD4+ T cells present in A 0/0 mice are sufficient for CM development. Neither the recently described CD4+, NK1.1+ T cell subset shown to be present in A 0/0 mice nor traditional NK cells seem to be required for the induction of CM because A 0/0 and C57BL/6 mice severely depleted of both NK1.1+ populations with mAb developed CM as readily as did normal Ig-treated controls. Deficiency of Th1-associated cytokines (IFN-gamma or IL-2) in mice by gene-targeted disruptions completely inhibited CM development, whereas the lack of Th2-associated cytokines (IL-4 or IL-10) did not prevent this disease. Our observation that B cell-deficient JHD and microMT mice developed CM provides evidence that neither B cells, their products, nor B cell Ag presentation are a requisite for CM pathology. We further observed that neither beta 2m 0/0 knockout mice, which lack CD8+ alpha beta T cells, nor C57BL/6 mice depleted of CD8+ T cells with anti-CD8 mAb treatment developed CM, leading us to conclude that CD8+ T cells are also crucial for the development of CM.
Article
Central nervous system (CNS) expression of two chemokine mRNAs, encoding monocyte chemoattractant protein-1 (MCP-1) and IFN-gamma-inducible protein (IP-10), was previously shown to be closely related to the onset of clinical signs of murine experimental autoimmune encephalomyelitis (EAE). Chemokine mRNAs accumulated in a striking, transient burst within astrocytes, near inflammatory leukocyte infiltrates. It remained unclear if chemokines functioned to initiate leukocyte entry into CNS tissues, or to amplify the intrathecal inflammatory reaction. To address this issue, we determined the expression of chemokine mRNAs at the earliest evidence of CNS immune-mediated inflammation. For these experiments, mice were sacrificed in pairs at varying times after immunization. Only one member of each pair was symptomatic for EAE at the time of sacrifice. Symptom presence correlated well with histological inflammation at the time of sacrifice. RNA was prepared from two CNS sites, brain and spinal cord, and expression of chemokine mRNAs was analyzed by a sensitive and quantitative reverse transcriptase/polymerase chain reaction dot-blot hybridization assay. CNS expressions of MCP-1 and IP-10 gene were correlated tightly with histological inflammation; indeed, chemokine expression was never detected in the absence of leukocyte infiltrates. In situ hybridizations showed that astrocytes expressed chemokine transcripts. These findings provide new information about mechanisms controlling chemokine mRNA expression during immune-mediated inflammation in EAE and are consistent with a role for chemokines as amplifiers of CNS inflammatory reactions.
Article
Infection with Plasmodium berghei ANKA (PbA) causes fatal cerebral malaria (CM). While a pathogenic role for tumor necrosis factor (TNF) has been established, we asked whether a disruption of interferon-gamma (IFN-gamma) signaling would modulate CM. We demonstrate here that IFN-gammaR-deficient mice are completely protected from CM. PbA-induced release of TNF and up-regulation of endothelial intercellular adhesion molecule (ICAM)-1 expression, recruitment of mononuclear cells, and cerebral microvascular damage with vascular leakage occur only in wild-type mice. Protected mice die at a later time of severe anemia and overwhelming parasitemia. Resistance to CM in IFN-gammaR-deficient mice is associated with reduced serum TNF levels, reduced interleukin-12 expression in the brain and increased T-helper 2 cytokines. In conclusion, IFN-gamma is apparently required for PbA-induced endothelial ICAM-1 up-regulation and subsequent microvascular pathology, resulting in fatal CM. In the absence of IFN-gamma signaling, ICAM-1 and TNF up-regulation is reduced; hence, PbA infection fails to cause fatal CM.
Article
Experimental autoimmune encephalomyelitis (EAE) is a T lymphocyte-mediated disease of the central nervous system (CNS), characterized by mononuclear cell infiltration and demyelination resulting in paralysis. We examined CC chemokine expression in the CNS throughout the entire course of the disease and found that the production of macrophage inflammatory protein (MIP)-1alpha correlated with increasing acute disease severity and remained elevated throughout chronic, relapsing disease. In contrast, a substantial level of monocyte chemotactic protein (MCP)-1 expression was not observed until late in acute disease and continued to be evident in the relapsing phase of the disease. MCP-1 expression correlated with increasing severity of clinical relapses. Lower levels of RANTES in the CNS were noted throughout the disease course, but showed little correlation with either acute or relapsing disease. Although RANTES expression was observed during the entire course of disease, anti-RANTES treatment had no effect on clinical disease progression. Anti-MCP-1, but not anti-MIP-1alpha, treatment during relapsing EAE decreased clinical severity of relapsing disease. Furthermore, anti-MCP-1 treatment reduced CNS macrophage accumulation during relapsing EAE. These results suggest that MIP-1alpha controls mononuclear cell accumulation during acute EAE, while MCP-1 controls mononuclear cell infiltration during relapsing EAE.
Article
Chemokines are low molecular weight chemotactic peptides that bind seven transmembrane-spanning, G protein-coupled receptors and deliver signals leading to T cell costimulation, hematopoeisis, cytokine expression, T cell differentiation, and integrin activation. Experimental autoimmune encephalomyelitis (EAE) is a CD4+ Th1-mediated demyelinating disease of the central nervous system (CNS) that serves as a model for multiple sclerosis (MS). A hallmark in the pathogenesis of this CNS demyelinating disease is the emigration of T cells and monocytes from the blood to the CNS. There are several considerations that suggest a role for chemokines in the influx of inflammatory cells and the resulting disease process including a tight temporal expression pattern with relationship to disease activity and prevention of disease development by in vivo neutralization. We review the evidence that temporal and spatial expressions of chemokines are crucial factors, complementing adhesion molecule upregulation, that regulate EAE and potentially MS disease activity as well as the functions of chemokines in Th1 and Th2 biology.
Article
To prevent uncontrolled expansion, the massive proliferation of T cells during an acute immune response has to be followed by controlled deletion. Here we show that similar to Fas, perforin is not only an important effector molecule of cytotoxic T lymphocytes (CTL) but also involved in down-regulating peripheral T cells. Mice deficient for both the CTL effector molecule perforin and the apoptosis-inducing Fas ligand spontaneously develop infiltration of highly activated CD8(+) T cells in kidney and liver and die between 5 and 12 weeks of age. Injection of staphylococcal enterotoxin B (SEB) into perforin-deficient mice results in dramatically increased selective expansion and prolonged persistence of CD8(+), but not CD4(+), SEB-reactive T cells. Also, secondary immunization of TCR transgenic perforin-deficient mice with the lymphocytic choriomeningitis virus glycoprotein-derived epitope peptide leads to an increased proliferation of transgenic CD8(+) T cells, that is not explained by failure to deplete professional antigen-presenting cells. These results point to a novel mechanism of T cell homeostasis in which the acquisition of perforin-dependent cytotoxic activity regulates the expansion and persistence of CD8(+) effector T cells in vivo.
Article
Malaria is a serious health, social and economic problem for over 40% of the world's population living in endemic regions. Of the half-billion people infected with malaria each year, some 2.5 million will develop cerebral complications. Even with expedient treatment with anti-malarials, the prognosis for an individual displaying symptoms of cerebral malaria remains poor, with an estimated 25%of cases resulting in death. There is, as yet, no direct treatment for cerebral malaria, and the exact mechanism by which it causes death is still undetermined.
Article
Since its introduction in 1994 (J. Immunol. Methods 171 (1994) 131), the flow cytometric analysis of lymphocyte proliferation by serial halving of the fluorescence intensity of the vital dye CFSE (carboxyfluorescein diacetate, succinimidyl ester or CFDA-SE) has become widely used in immunological laboratories around the world. This technique allows the visualisation of eight to 10 discrete cycles of cell division by flow cytometry, both in vitro and in vivo. Appropriately conjugated antibodies can be used to probe surface marker changes as cells divide, or changes in expression of internal molecules such as cytokines when appropriate fixation and permeabilisation protocols are used. An added advantage of the technique is the ability to recover viable cells which have undergone defined numbers of cell divisions by flow cytometric sorting, allowing functional studies to be performed. Other commonly used assays of cell proliferation give only limited information, as they usually measure division at a population level. The CFSE technique can be used to determine kinetics of immune responses, track proliferation in minor subsets of cells and follow the acquisition of differentiation markers or internal proteins linked to cell division.
Article
T cell memory depends on factors that regulate expansion and death of these cells after antigenic stimulation. Mice deficient in perforin and interferon-gamma (IFN-gamma) exhibited increased expansion, altered immunodominance, and decreased death of antigen-specific CD8+ T cells after infection with an attenuated strain of Listeria monocytogenes, which was cleared from these mice. Expansion of CD8+ T cells was controlled by perforin, whereas IFN-gamma regulated immunodominance and the death phase. Thus, perforin and IFN-gamma regulate distinct elements of CD8+ T cell homeostasis independently of their role as antimicrobial effector molecules.
Article
We examine here several diseases that are associated with inappropriate activation of the chemokine network. Detailed comment has been restricted to pathological states for which there are compelling data either from clinical observations or animal models. These include cardiovascular disease, allergic inflammatory disease, transplantation, neuroinflammation, cancer and HIV-associated disease. Discussion focuses on therapeutic directions in which the rapidly evolving chemokine field appears to be headed.
Article
Tissue-selective trafficking of memory and effector T and B lymphocytes is mediated by unique combinations of adhesion molecules and chemokines. The discovery of several related epithelial-expressed chemokines (TECK/CCL25 in small intestine, CTACK/CCL27 in skin, and MEC/CCL28 in diverse mucosal sites) now highlights an important role for epithelial cells in controlling homeostatic lymphocyte trafficking, including the localization of cutaneous and intestinal memory T cells, and of IgA plasma cells. Constitutively expressed epithelial chemokines may help determine the character of local immune responses and contribute to the systemic organization of the immune system.
Article
Different diseases of the CNS are associated with blood-brain barrier (BBB) damage and mononuclear cell infiltration. In order to study genes that may play a role in endothelial cell regulation in inflammatory CNS diseases, we performed differential gene expression (DGE) analysis using a mouse brain endothelial cell line. We found that interferon-gamma (IFNgamma)-induced monokine (MIG), a chemokine that plays a role in T lymphocyte and monocyte chemoattraction, is highly expressed in the presence of inflammatory cytokines. We show that MIG, produced by brain endothelial cells in vitro, is biologically active in attractingT lymphocytes and that it is possible to interfere with this mechanism of action using anti-MIG antibodies. We suggest that blocking MIG may be beneficial in CNS inflammation. We detected constitutive expression of the MIG receptor, CXCR3, on the surface of the endothelial cells and therefore hypothesize that it plays a role in maintaining the cytokine gradient at the region of CNS inflammation.
Article
Fractalkine (FKN), also known as neurotactin, is a CX(3)C chemokine that exists in both secreted and neuronal membrane-bound forms and is upregulated during brain inflammation. There is accumulating evidence that FKN induces chemotaxis by binding to its receptor CX(3)CR1 on leukocytes and microglia. We generated FKN-deficient mice to study the role of FKN in postischemic brain injury. After transient focal cerebral ischemia, FKN-deficient mice had a 28% reduction in infarction size and lower mortality rate, when compared to wild-type littermates. The findings of this study indicate a possible role for FKN in augmenting postischemic injury and mortality after transient focal cerebral ischemia.
Article
Leukocyte subsets are recruited from the blood to lymphoid and non-lymphoid tissues via a multi-step process that involves distinct adhesive and activation steps. Chemokines, a family of chemotactic cytokines that signal through G-protein-coupled receptors, play critical roles in regulating the leukocyte recruitment cascade. Chemokines can be transported and immobilized on the surface of vascular endothelial cells, where they activate leukocyte subsets expressing specific receptors. Activation signals induce firm adhesion of rolling leukocytes by rapidly upregulating integrin affinity and/or avidity. Chemokines can also direct migration of adherent cells across the endothelium, and control segregation of cells into specific microenvironments within tissues. The regulated expression of chemokines and their receptors is a critical determinant for homing of specialized lymphocyte subsets, and controls both tissue and inflammation-specific immune processes.
Article
Experimental autoimmune encephalomyelitis (EAE) is a T cell mediated demyelinating disease of the central nervous system (CNS) that serves as a model for multiple sclerosis (MS). Insights into the pathogenesis of this model may help scientists understand the human disease and aid in rational drug discovery. In this review we summarize the role of chemokines and chemokine receptors in disease pathogenesis and suggest a pathway of events that leads to demyelination and subsequent clinical disease manifestation.
The roles of perforin-and Fas-dependent cytotoxicity in protection against cytopathic and noncytopathic viruses http://www.jimmunol.org/ Downloaded from 19 Molecular mechanisms of sequestration in malaria
  • D Kagi
  • P Seiler
  • J Pavlovic
  • B Ledermann
  • K Burki
  • R M Zinkernagel
  • H Hengartner
Kagi, D., P. Seiler, J. Pavlovic, B. Ledermann, K. Burki, R. M. Zinkernagel, and H. Hengartner. 1995. The roles of perforin-and Fas-dependent cytotoxicity in protection against cytopathic and noncytopathic viruses. Eur. J. Immunol. 25: 3256. 2227 The Journal of Immunology by guest on June 9, 2013 http://www.jimmunol.org/ Downloaded from 19. Berendt, A. R., D. J. Ferguson, J. Gardner, G. Turner, A. Rowe, C. McCormick, D. Roberts, A. Craig, R. Pinches, B. C. Elford, et al. 1994. Molecular mechanisms of sequestration in malaria. Parasitology 108:S19.
Molecular mechanisms of lymphocyte-mediated cytotoxicity and their role in immunological protection and pathogenesis in vivo
  • D Kagi
  • K B. Ledermann
  • R M Burki
  • H Zinkernagel
  • Hengartner
Kagi, D., B. Ledermann, K. Burki, R. M. Zinkernagel, and H. Hengartner. 1996. Molecular mechanisms of lymphocyte-mediated cytotoxicity and their role in immunological protection and pathogenesis in vivo. Annu. Rev. Immunol. 14:207.
World Health Organization, Division of Control of Tropical Diseases Severe and complicated malaria
World Health Organization, Division of Control of Tropical Diseases. 1990. Severe and complicated malaria. Trans. R. Soc. Trop. Med. Hyg. 84:1.
Mice deficient in fractalkine are less susceptible to cerebral ischemia-reperfusion injury
  • S G Soriano
  • L S Amaravadi
  • Y F Wang
  • H Zhou
  • G X Yu
  • J R Tonra
  • V Fairchild-Huntress
  • Q Fang
  • J H Dunmore
  • D Huszar
Soriano, S. G., L. S. Amaravadi, Y. F. Wang, H. Zhou, G. X. Yu, J. R. Tonra, V. Fairchild-Huntress, Q. Fang, J. H. Dunmore, D. Huszar, et al. 2002. Mice deficient in fractalkine are less susceptible to cerebral ischemia-reperfusion injury. J. Neuroimmunol. 125:59.
World Health Organization Division of Control of Tropical Diseases