Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.080
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 18(1): 54-63, 2017 01.
Article in English | MEDLINE | ID: mdl-27721430

ABSTRACT

Genes and pathways in which inactivation dampens tissue inflammation present new opportunities for understanding the pathogenesis of common human inflammatory diseases, including inflammatory bowel disease, rheumatoid arthritis and multiple sclerosis. We identified a mutation in the gene encoding the deubiquitination enzyme USP15 (Usp15L749R) that protected mice against both experimental cerebral malaria (ECM) induced by Plasmodium berghei and experimental autoimmune encephalomyelitis (EAE). Combining immunophenotyping and RNA sequencing in brain (ECM) and spinal cord (EAE) revealed that Usp15L749R-associated resistance to neuroinflammation was linked to dampened type I interferon responses in situ. In hematopoietic cells and in resident brain cells, USP15 was coexpressed with, and functionally acted together with the E3 ubiquitin ligase TRIM25 to positively regulate type I interferon responses and to promote pathogenesis during neuroinflammation. The USP15-TRIM25 dyad might be a potential target for intervention in acute or chronic states of neuroinflammation.


Subject(s)
DNA-Binding Proteins/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Malaria, Cerebral/immunology , Neurogenic Inflammation/immunology , Transcription Factors/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , DNA-Binding Proteins/genetics , Encephalomyelitis, Autoimmune, Experimental/drug therapy , HEK293 Cells , Humans , Immunity, Innate , Interferon Type I/metabolism , Malaria, Cerebral/drug therapy , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Molecular Targeted Therapy , Myelin-Oligodendrocyte Glycoprotein/immunology , Neurogenic Inflammation/drug therapy , Peptide Fragments/immunology , Plasmodium berghei/immunology , Transcription Factors/genetics , Ubiquitin-Specific Proteases/genetics
2.
Nature ; 611(7936): 563-569, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36352220

ABSTRACT

Malaria infection involves an obligatory, yet clinically silent liver stage1,2. Hepatocytes operate in repeating units termed lobules, exhibiting heterogeneous gene expression patterns along the lobule axis3, but the effects of hepatocyte zonation on parasite development at the molecular level remain unknown. Here we combine single-cell RNA sequencing4 and single-molecule transcript imaging5 to characterize the host and parasite temporal expression programmes in a zonally controlled manner for the rodent malaria parasite Plasmodium berghei ANKA. We identify differences in parasite gene expression in distinct zones, including potentially co-adaptive programmes related to iron and fatty acid metabolism. We find that parasites develop more rapidly in the pericentral lobule zones and identify a subpopulation of periportally biased hepatocytes that harbour abortive infections, reduced levels of Plasmodium transcripts and parasitophorous vacuole breakdown. These 'abortive hepatocytes', which appear predominantly with high parasite inoculum, upregulate immune recruitment and key signalling programmes. Our study provides a resource for understanding the liver stage of Plasmodium infection at high spatial resolution and highlights the heterogeneous behaviour of both the parasite and the host hepatocyte.


Subject(s)
Gene Expression Regulation , Hepatocytes , Liver , Malaria , Parasites , Plasmodium berghei , Single-Cell Analysis , Animals , Hepatocytes/cytology , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/parasitology , Liver/anatomy & histology , Liver/cytology , Liver/immunology , Liver/parasitology , Malaria/genetics , Malaria/immunology , Malaria/parasitology , Parasites/genetics , Parasites/immunology , Parasites/metabolism , Plasmodium berghei/genetics , Plasmodium berghei/immunology , Plasmodium berghei/metabolism , Single Molecule Imaging , Sequence Analysis, RNA , Iron/metabolism , Fatty Acids/metabolism , Transcription, Genetic , Genes, Protozoan/genetics , Host-Parasite Interactions/genetics , Host-Parasite Interactions/immunology
3.
Immunity ; 48(4): 760-772.e4, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29625893

ABSTRACT

Cerebral malaria is a deadly complication of Plasmodium infection and involves blood brain barrier (BBB) disruption following infiltration of white blood cells. During experimental cerebral malaria (ECM), mice inoculated with Plasmodium berghei ANKA-infected red blood cells develop a fatal CM-like disease caused by CD8+ T cell-mediated pathology. We found that treatment with interleukin-15 complex (IL-15C) prevented ECM, whereas IL-2C treatment had no effect. IL-15C-expanded natural killer (NK) cells were necessary and sufficient for protection against ECM. IL-15C treatment also decreased CD8+ T cell activation in the brain and prevented BBB breakdown without influencing parasite load. IL-15C induced NK cells to express IL-10, which was required for IL-15C-mediated protection against ECM. Finally, we show that ALT-803, a modified human IL-15C, mediates similar induction of IL-10 in NK cells and protection against ECM. These data identify a regulatory role for cytokine-stimulated NK cells in the prevention of a pathogenic immune response.


Subject(s)
Interleukin-10/immunology , Interleukin-15/immunology , Killer Cells, Natural/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Proteins/pharmacology , Animals , Blood-Brain Barrier/pathology , Brain/immunology , Brain/pathology , CD8-Positive T-Lymphocytes/immunology , Interleukin-10/biosynthesis , Lymphocyte Activation/immunology , Malaria, Cerebral/microbiology , Malaria, Cerebral/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Recombinant Fusion Proteins
4.
Immunity ; 47(5): 835-847.e4, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29150238

ABSTRACT

Immune response (Ir) genes, originally proposed by Baruj Benacerraf to explain differential antigen-specific responses in animal models, have become synonymous with the major histocompatibility complex (MHC). We discovered a non-MHC-linked Ir gene in a T cell receptor (TCR) locus that was required for CD8+ T cell responses to the Plasmodium berghei GAP5040-48 epitope in mice expressing the MHC class I allele H-2Db. GAP5040-48-specific CD8+ T cell responses emerged from a very large pool of naive Vß8.1+ precursors, which dictated susceptibility to cerebral malaria and conferred protection against recombinant Listeria monocytogenes infection. Structural analysis of a prototypical Vß8.1+ TCR-H-2Db-GAP5040-48 ternary complex revealed that germline-encoded complementarity-determining region 1ß residues present exclusively in the Vß8.1 segment mediated essential interactions with the GAP5040-48 peptide. Collectively, these findings demonstrated that Vß8.1 functioned as an Ir gene that was indispensable for immune reactivity against the malaria GAP5040-48 epitope.


Subject(s)
Histocompatibility Antigen H-2D/genetics , Plasmodium berghei/immunology , Protozoan Proteins/immunology , Receptors, Antigen, T-Cell/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , Complementarity Determining Regions , Epitopes , Genes, T-Cell Receptor beta , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptide Fragments/immunology
5.
Immunity ; 47(6): 1197-1209.e10, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29195810

ABSTRACT

Antibodies against the NANP repeat of circumsporozoite protein (CSP), the major surface antigen of Plasmodium falciparum (Pf) sporozoites, can protect from malaria in animal models but protective humoral immunity is difficult to induce in humans. Here we cloned and characterized rare affinity-matured human NANP-reactive memory B cell antibodies elicited by natural Pf exposure that potently inhibited parasite transmission and development in vivo. We unveiled the molecular details of antibody binding to two distinct protective epitopes within the NANP repeat. NANP repeat recognition was largely mediated by germline encoded and immunoglobulin (Ig) heavy-chain complementarity determining region 3 (HCDR3) residues, whereas affinity maturation contributed predominantly to stabilizing the antigen-binding site conformation. Combined, our findings illustrate the power of exploring human anti-CSP antibody responses to develop tools for malaria control in the mammalian and the mosquito vector and provide a molecular basis for the structure-based design of next-generation CSP malaria vaccines.


Subject(s)
Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Immunity, Humoral , Immunoglobulin Heavy Chains/immunology , Malaria, Falciparum/prevention & control , Protozoan Proteins/immunology , Animals , Antibodies, Protozoan/biosynthesis , Antibodies, Protozoan/chemistry , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , B-Lymphocytes/immunology , B-Lymphocytes/parasitology , Crystallography, X-Ray , Epitopes/chemistry , Epitopes/immunology , Female , Gene Expression , Humans , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Heavy Chains/chemistry , Immunologic Memory , Malaria/immunology , Malaria/parasitology , Malaria/prevention & control , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Male , Mice , Models, Molecular , Plasmodium berghei/immunology , Plasmodium falciparum/immunology , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sporozoites/chemistry , Sporozoites/immunology
6.
Immunity ; 44(3): 672-682, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26968425

ABSTRACT

Interleukin-27 (IL-27) is a heterodimeric regulatory cytokine of the IL-12 family, which is produced by macrophages, dendritic cells, and B cells upon stimulation through innate immune receptors. Here, we described regulatory CD4(+) T cells that produce IL-27 in response to T cell receptor stimulation during malaria infection, inhibiting IL-2 production and clonal expansion of other T cells in an IL-27-dependent manner. IL-27-producing CD4(+) T cells were Foxp3(-)CD11a(+)CD49d(+) malaria antigen-specific CD4(+) T cells and were distinct from interferon-γ (IFN-γ) producing Th1 or IL-10 producing Tr1 cells. In mice lacking IL-27 in T cells, IL-2 production was restored and clonal expansion and IFN-γ production by specific CD4(+) T cells were improved, culminating in reduced parasite burden. This study highlights a unique population of IL-27 producing regulatory CD4(+) T cells and their critical role in the regulation of the protective immune response against malaria parasites.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Interleukin-27/metabolism , Malaria/immunology , Plasmodium berghei/immunology , T-Lymphocytes, Regulatory/physiology , Animals , CD4-Positive T-Lymphocytes/parasitology , Cell Proliferation/genetics , Cells, Cultured , Cytokines/metabolism , Immunity, Innate , Interleukin-27/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell/metabolism , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Receptors, Interleukin , T-Lymphocytes, Regulatory/parasitology
7.
Immunity ; 45(4): 889-902, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27692609

ABSTRACT

In recent years, various intervention strategies have reduced malaria morbidity and mortality, but further improvements probably depend upon development of a broadly protective vaccine. To better understand immune requirement for protection, we examined liver-stage immunity after vaccination with irradiated sporozoites, an effective though logistically difficult vaccine. We identified a population of memory CD8+ T cells that expressed the gene signature of tissue-resident memory T (Trm) cells and remained permanently within the liver, where they patrolled the sinusoids. Exploring the requirements for liver Trm cell induction, we showed that by combining dendritic cell-targeted priming with liver inflammation and antigen recognition on hepatocytes, high frequencies of Trm cells could be induced and these cells were essential for protection against malaria sporozoite challenge. Our study highlights the immune potential of liver Trm cells and provides approaches for their selective transfer, expansion, or depletion, which may be harnessed to control liver infections or autoimmunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Liver/immunology , Malaria/immunology , Animals , CD8-Positive T-Lymphocytes/parasitology , Culicidae , Dendritic Cells/immunology , Dendritic Cells/parasitology , Hepatocytes/immunology , Hepatocytes/parasitology , Liver/parasitology , Liver Diseases/immunology , Liver Diseases/parasitology , Malaria Vaccines/immunology , Mice , Plasmodium berghei/immunology , Sporozoites/immunology , Sporozoites/parasitology , Vaccination/methods
8.
Proc Natl Acad Sci U S A ; 119(35): e2209729119, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35994647

ABSTRACT

Glutaminyl cyclase (QC) modifies N-terminal glutamine or glutamic acid residues of target proteins into cyclic pyroglutamic acid (pGlu). Here, we report the biochemical and functional analysis of Plasmodium QC. We show that sporozoites of QC-null mutants of rodent and human malaria parasites are recognized by the mosquito immune system and melanized when they reach the hemocoel. Detailed analyses of rodent malaria QC-null mutants showed that sporozoite numbers in salivary glands are reduced in mosquitoes infected with QC-null or QC catalytically dead mutants. This phenotype can be rescued by genetic complementation or by disrupting mosquito melanization or phagocytosis by hemocytes. Mutation of a single QC-target glutamine of the major sporozoite surface protein (circumsporozoite protein; CSP) of the rodent parasite Plasmodium berghei also results in melanization of sporozoites. These findings indicate that QC-mediated posttranslational modification of surface proteins underlies evasion of killing of sporozoites by the mosquito immune system.


Subject(s)
Aminoacyltransferases , Culicidae , Malaria , Protein Processing, Post-Translational , Sporozoites , Aminoacyltransferases/immunology , Animals , Culicidae/immunology , Glutamic Acid/metabolism , Glutamine/metabolism , Humans , Malaria/genetics , Malaria/immunology , Malaria/parasitology , Plasmodium berghei/genetics , Plasmodium berghei/immunology , Protein Processing, Post-Translational/immunology , Protozoan Proteins/immunology , Sporozoites/immunology
9.
J Infect Dis ; 229(6): 1894-1903, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38408353

ABSTRACT

BACKGROUND: Plasmodium falciparum and Plasmodium vivax account for >90% global malaria burden. Transmission intervention strategies encompassing transmission-blocking vaccines (TBV) and drugs represent ideal public health tools to eliminate malaria at the population level. The availability of mature P. falciparum gametocytes through in vitro culture has facilitated development of a standard membrane feeding assay to assess efficacy of transmission interventions against P. falciparum. The lack of in vitro culture for P. vivax has significantly hampered similar progress on P. vivax and limited studies have been possible using blood from infected patients in endemic areas. The ethical and logistical limitations of on-time access to blood from patients have impeded the development of P. vivax TBVs. METHODS: Transgenic murine malaria parasites (Plasmodium berghei) expressing TBV candidates offer a promising alternative for evaluation of P. vivax TBVs through in vivo studies in mice, and ex vivo membrane feeding assay (MFA). RESULTS: We describe the development of transmission-competent transgenic TgPbvs25 parasites and optimization of parameters to establish an ex vivo MFA to evaluate P. vivax TBV based on Pvs25 antigen. CONCLUSIONS: The MFA is expected to expedite Pvs25-based TBV development without dependence on blood from P. vivax-infected patients in endemic areas for evaluation.


Subject(s)
Malaria Vaccines , Malaria, Vivax , Plasmodium berghei , Plasmodium vivax , Animals , Malaria Vaccines/immunology , Malaria Vaccines/genetics , Plasmodium vivax/genetics , Plasmodium vivax/immunology , Malaria, Vivax/transmission , Malaria, Vivax/prevention & control , Malaria, Vivax/parasitology , Plasmodium berghei/genetics , Plasmodium berghei/immunology , Mice , Antigens, Protozoan/immunology , Antigens, Protozoan/genetics , Humans , Female , Antigens, Surface
10.
J Immunol ; 208(5): 1292-1304, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35131868

ABSTRACT

Pathogen-specific CD8 T cells face the problem of finding rare cells that present their cognate Ag either in the lymph node or in infected tissue. Although quantitative details of T cell movement strategies in some tissues such as lymph nodes or skin have been relatively well characterized, we still lack quantitative understanding of T cell movement in many other important tissues, such as the spleen, lung, liver, and gut. We developed a protocol to generate stable numbers of liver-located CD8 T cells, used intravital microscopy to record movement patterns of CD8 T cells in livers of live mice, and analyzed these and previously published data using well-established statistical and computational methods. We show that, in most of our experiments, Plasmodium-specific liver-localized CD8 T cells perform correlated random walks characterized by transiently superdiffusive displacement with persistence times of 10-15 min that exceed those observed for T cells in lymph nodes. Liver-localized CD8 T cells typically crawl on the luminal side of liver sinusoids (i.e., are in the blood); simulating T cell movement in digital structures derived from the liver sinusoids illustrates that liver structure alone is sufficient to explain the relatively long superdiffusive displacement of T cells. In experiments when CD8 T cells in the liver poorly attach to the sinusoids (e.g., 1 wk after immunization with radiation-attenuated Plasmodium sporozoites), T cells also undergo Lévy flights: large displacements occurring due to cells detaching from the endothelium, floating with the blood flow, and reattaching at another location. Our analysis thus provides quantitative details of movement patterns of liver-localized CD8 T cells and illustrates how structural and physiological details of the tissue may impact T cell movement patterns.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Movement/physiology , Liver/immunology , Malaria/prevention & control , Plasmodium berghei/immunology , Animals , Capillaries/cytology , Cellular Microenvironment/physiology , Liver/blood supply , Malaria/pathology , Mice , Plasmodium berghei/growth & development , Sporozoites/growth & development , Sporozoites/immunology , Vaccination
11.
Eur J Immunol ; 52(2): 312-327, 2022 02.
Article in English | MEDLINE | ID: mdl-34752634

ABSTRACT

Overwhelming activation of T cells in acute malaria is associated with severe outcomes. Thus, counter-regulation by anti-inflammatory mechanisms is indispensable for an optimal resolution of disease. Using Plasmodium berghei ANKA (PbA) infection of C57BL/6 mice, we performed a comprehensive analysis of co-inhibitory molecules expressed on CD4+ and CD8+ T cells using an unbiased cluster analysis approach. We identified similar T cell clusters co-expressing several co-inhibitory molecules like programmed cell death protein 1 (PD-1) and lymphocyte activation gene 3 (LAG-3) in the CD4+ and the CD8+ T cell compartment. Interestingly, despite expressing co-inhibitory molecules, which are associated with T cell exhaustion in chronic settings, these T cells were more functional compared to activated T cells that were negative for co-inhibitory molecules. However, T cells expressing high levels of PD-1 and LAG-3 also conferred suppressive capacity and thus resembled type I regulatory T cells. To our knowledge, this is the first description of malaria-induced CD8+ T cells with suppressive capacity. Importantly, we found an induction of T cells with a similar co-inhibitory rich phenotype in Plasmodium falciparum-infected patients. In conclusion, we demonstrate that malaria-induced T cells expressing co-inhibitory molecules are not exhausted, but acquire additional suppressive capacity, which might represent an immune regulatory pathway to prevent further activation of T cells during acute malaria.


Subject(s)
Antigens, CD/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Gene Expression Regulation/immunology , Immune Tolerance , Malaria, Falciparum/immunology , Plasmodium berghei/immunology , Plasmodium falciparum/immunology , Programmed Cell Death 1 Receptor/immunology , Adolescent , Adult , Animals , Female , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Lymphocyte Activation Gene 3 Protein
12.
Eur J Immunol ; 52(2): 270-284, 2022 02.
Article in English | MEDLINE | ID: mdl-34773640

ABSTRACT

Recognition of pathogen-associated molecular patterns (PAMPs) through Toll-like receptors (TLRs) plays a pivotal role in first-line pathogen defense. TLRs are also likely triggered during a Plasmodium infection in vivo by parasite-derived components. However, the contribution of innate responses to liver infection and to the subsequent clinical outcome of a blood infection is not well understood. To assess the potential effects of enhanced TLR-signalling on Plasmodium infection, we systematically examined the effect of agonist-primed immune responses to sporozoite inoculation in the P. berghei/C57Bl/6 murine malaria model. We could identify distinct stage-specific effects on the course of infection after stimulation with two out of four TLR-ligands tested. Priming with a TLR9 agonist induced killing of pre-erythrocytic stages in the liver that depended on macrophages and the expression of inducible nitric oxide synthase (iNOS). These factors have previously not been recognized as antigen-independent effector mechanisms against Plasmodium liver stages. Priming with TLR4 and -9 agonists also translated into blood stage-specific protection against experimental cerebral malaria (ECM). These insights are relevant to the activation of TLR signalling pathways by adjuvant systems of antimalaria vaccine strategies. The protective role of TLR4-activation against ECM might also explain some unexpected clinical effects observed with pre-erythrocytic vaccine approaches.


Subject(s)
Liver Diseases , Liver , Macrophage Activation , Macrophages/immunology , Malaria , Plasmodium berghei/immunology , Signal Transduction , Toll-Like Receptor 9/immunology , Animals , Female , Liver/immunology , Liver/parasitology , Liver Diseases/genetics , Liver Diseases/immunology , Liver Diseases/parasitology , Malaria/genetics , Malaria/immunology , Mice , Mice, Transgenic , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 9/genetics
13.
Int Immunol ; 34(1): 21-33, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34648636

ABSTRACT

Plasmodium parasites that infect humans are highly polymorphic, and induce various infections ranging from an asymptomatic state to life-threatening diseases. However, how the differences between the parasites affect host immune responses during blood-stage infection remains largely unknown. We investigated the CD4+ T-cell immune responses in mice infected with P. berghei ANKA (PbA) or P. chabaudi chabaudi AS (Pcc) using PbT-II cells, which recognize a common epitope of these parasites. In the acute phase of infection, CD4+ T-cell responses in PbA-infected mice showed a lower involvement of Th1 cells and a lower proportion of Ly6Clo effector CD4+ T cells than those in Pcc-infected mice. Transcriptome analysis of PbT-II cells indicated that type I interferon (IFN)-regulated genes were expressed at higher levels in both Th1- and Tfh-type PbT-II cells from PbA-infected mice than those from Pcc-infected mice. Moreover, IFN-α levels were considerably higher in PbA-infected mice than in Pcc-infected mice. Inhibition of type I IFN signaling increased PbT-II and partially reversed the Th1 over Tfh bias of the PbT-II cells in both PbA- and Pcc-infected mice. In the memory phase, PbT-II cells in PbA-primed mice maintained higher numbers and exhibited a better recall response to the antigen. However, recall responses were not significantly different between the infection groups after re-challenge with PbA, suggesting the effect of the inflammatory environment by the infection. These observations suggest that the differences in Plasmodium-specific CD4+ T-cell responses between PbA- and Pcc-infected mice were associated with the difference in type I IFN production during the early phase of the infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interferon Type I/biosynthesis , Malaria/immunology , Plasmodium berghei/immunology , Plasmodium chabaudi/immunology , Animals , Cells, Cultured , Mice , Mice, Transgenic
14.
J Immunol ; 207(6): 1578-1590, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34400523

ABSTRACT

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.


Subject(s)
Blood-Brain Barrier/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Adoptive Transfer/methods , Animals , Disease Models, Animal , Female , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Parasitemia/immunology , Spleen/immunology
15.
J Biol Chem ; 296: 100615, 2021.
Article in English | MEDLINE | ID: mdl-33798555

ABSTRACT

Persistent high levels of proinflammatory and Th1 responses contribute to cerebral malaria (CM). Suppression of inflammatory responses and promotion of Th2 responses prevent pathogenesis. IL-4 commonly promotes Th2 responses and inhibits inflammatory and Th1 responses. Therefore, IL-4 is widely considered as a beneficial cytokine via its Th2-promoting role that is predicted to provide protection against severe malaria by inhibiting inflammatory responses. However, IL-4 may also induce inflammatory responses, as the result of IL-4 action depends on the timing and levels of its production and the tissue environment in which it is produced. Recently, we showed that dendritic cells (DCs) produce IL-4 early during malaria infection in response to a parasite protein and that this IL-4 response may contribute to severe malaria. However, the mechanism by which IL-4 produced by DCs contributing to lethal malaria is unknown. Using Plasmodium berghei ANKA-infected C57BL/6 mice, a CM model, we show here that mice lacking IL-4Rα only in CD8α+ DCs are protected against CM pathogenesis and survive, whereas WT mice develop CM and die. Compared with WT mice, mice lacking IL-4Rα in CD11c+ or CD8α+ DCs showed reduced inflammatory responses leading to decreased Th1 and cytotoxic CD8+ T cell responses, lower infiltration of CD8+ T cells to the brain, and negligible brain pathology. The novel results presented here reveal a paradoxical role of IL-4Rα signaling in CM pathogenesis that promotes CD8α+ DC-mediated inflammatory responses that generate damaging Th1 and cytotoxic CD8+ T cell responses.


Subject(s)
CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Receptors, Cell Surface/immunology , Signal Transduction/immunology , Th1 Cells/immunology , Animals , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/pathology , Dendritic Cells/pathology , Interleukin-4/genetics , Interleukin-4/immunology , Malaria, Cerebral/genetics , Malaria, Cerebral/pathology , Mice , Mice, Knockout , Plasmodium berghei/genetics , Receptors, Cell Surface/genetics , Signal Transduction/genetics , Th1 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology
16.
Infect Immun ; 90(1): e0035921, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34724388

ABSTRACT

Malaria begins when an infected mosquito injects saliva containing Plasmodium sporozoites into the skin of a vertebrate host. Passive immunization of mice with antiserum against the Anopheles gambiae mosquito saliva protein TRIO (AgTRIO) offers significant protection against Plasmodium infection of mice. Furthermore, passive transfer of both AgTRIO antiserum and an anti-circumsporozoite protein monoclonal antibody provides synergistic protection. In this study, we generated monoclonal antibodies against AgTRIO to delineate the regions of AgTRIO associated with protective immunity. Monoclonal antibody 13F-1 markedly reduced Plasmodium infection in mice and recognized a region (VDDLMAKFN) in the carboxyl terminus of AgTRIO. 13F-1 is an IgG2a isotype monoclonal antibody, and the Fc region is required for protection. These data will aid in the generation of future malaria vaccines that may include both pathogen and vector antigens.


Subject(s)
Anopheles/immunology , Antibodies, Monoclonal/immunology , Culicidae/immunology , Malaria/immunology , Malaria/prevention & control , Amino Acid Sequence , Animals , Disease Models, Animal , Immunization, Passive , Immunoglobulin Fc Fragments , Insect Proteins/chemistry , Insect Proteins/immunology , Malaria/parasitology , Mice , Plasmodium berghei/immunology , Protein Binding/immunology , Protein Interaction Domains and Motifs/immunology
17.
PLoS Pathog ; 16(9): e1008799, 2020 09.
Article in English | MEDLINE | ID: mdl-32898164

ABSTRACT

Professional antigen-presenting cells (APCs), like macrophages (Mϕs) and dendritic cells (DCs), are central players in the induction of natural and vaccine-induced immunity to malaria, yet very little is known about the interaction of SPZ with human APCs. Intradermal delivery of whole-sporozoite vaccines reduces their effectivity, possibly due to dermal immunoregulatory effects. Therefore, understanding these interactions could prove pivotal to malaria vaccination. We investigated human APC responses to recombinant circumsporozoite protein (recCSP), SPZ and anti-CSP opsonized SPZ both in monocyte derived MoDCs and MoMϕs. Both MoDCs and MoMϕs readily took up recCSP but did not change phenotype or function upon doing so. SPZ are preferentially phagocytosed by MoMϕs instead of DCs and phagocytosis greatly increased after opsonization. Subsequently MoMϕs show increased surface marker expression of activation markers as well as tolerogenic markers such as Programmed Death-Ligand 1 (PD-L1). Additionally they show reduced motility, produce interleukin 10 and suppressed interferon gamma (IFNγ) production by antigen specific CD8+ T cells. Importantly, we investigated phenotypic responses to SPZ in primary dermal APCs isolated from human skin explants, which respond similarly to their monocyte-derived counterparts. These findings are a first step in enhancing our understanding of pre-erythrocytic natural immunity and the pitfalls of intradermal vaccination-induced immunity.


Subject(s)
Antigen-Presenting Cells/immunology , Macrophages/immunology , Malaria/immunology , Plasmodium berghei/immunology , Protozoan Proteins/immunology , Skin/immunology , Sporozoites/immunology , Animals , Cells, Cultured , Female , Humans , Macrophages/parasitology , Malaria/parasitology , Mice , Skin/parasitology
18.
PLoS Pathog ; 16(10): e1008985, 2020 10.
Article in English | MEDLINE | ID: mdl-33045027

ABSTRACT

The arthropod melanization immune response is activated by extracellular protease cascades predominantly comprised of CLIP-domain serine proteases (CLIP-SPs) and serine protease homologs (CLIP-SPHs). In the malaria vector, Anopheles gambiae, the CLIP-SPHs SPCLIP1, CLIPA8, and CLIPA28 form the core of a hierarchical cascade downstream of mosquito complement that is required for microbial melanization. However, our understanding of the regulatory relationship of the CLIP-SPH cascade with the catalytic CLIP-SPs driving melanization is incomplete. Here, we report on the development of a novel screen to identify melanization pathway components based on the quantitation of melanotic mosquito excreta, eliminating the need for microdissections or hemolymph enzymatic assays. Using this screen, we identified CLIPC9 and subsequent functional analyses established that this protease is essential for the melanization of both Escherichia coli and the rodent malaria parasite Plasmodium berghei. Mechanistically, septic infection with E. coli promotes CLIPC9 cleavage and both full-length and cleaved CLIPC9 localize to this bacterium in a CLIPA8-dependent manner. The steady state level of CLIPC9 in the hemolymph is regulated by thioester-containing protein 1 (TEP1), suggesting it functions downstream of mosquito complement. In support, CLIPC9 cleavage is inhibited following SPCLIP1, CLIPA8, and CLIPA28 knockdown positioning it downstream of the CLIP-SPH cascade. Moreover, like CLIPA8 and CLIPA28, CLIPC9 processing is negatively regulated by serine protease inhibitor 2 (SRPN2). This report demonstrates how our novel excretion-based approach can be utilized to dissect the complex protease networks regulating mosquito melanization. Collectively, our findings establish that CLIPC9 is required for microbial melanization in An. gambiae and shed light on how the CLIP-SPH cascade regulates this potent immune response.


Subject(s)
Anopheles/parasitology , Insect Proteins/metabolism , Malaria/parasitology , Melanins/metabolism , Mosquito Vectors/parasitology , Serine Proteases/metabolism , Serine/metabolism , Animals , Anopheles/immunology , Insect Proteins/genetics , Malaria/immunology , Malaria/metabolism , Malaria/pathology , Mice , Plasmodium berghei/immunology , Plasmodium berghei/isolation & purification
19.
PLoS Pathog ; 16(4): e1008453, 2020 04.
Article in English | MEDLINE | ID: mdl-32330198

ABSTRACT

Malaria, caused by the protozoan parasite Plasmodium and transmitted by Anopheles mosquitoes, represents a major threat to human health. Plasmodium's infection cycle in the Anopheles vector is critical for transmission of the parasite between humans. The midgut-stage bottleneck of infection is largely imposed by the mosquito's innate immune system. microRNAs (miRNAs, small noncoding RNAs that bind to target RNAs to regulate gene expression) are also involved in regulating immunity and the anti-Plasmodium defense in mosquitoes. Here, we characterized the mosquito's miRNA responses to Plasmodium infection using an improved crosslinking and immunoprecipitation (CLIP) method, termed covalent ligation of endogenous Argonaute-bound RNAs (CLEAR)-CLIP. Three candidate miRNAs' influence on P. falciparum infection and midgut microbiota was studied through transgenically expressed miRNA sponges (miR-SPs) in midgut and fat body tissues. MiR-SPs mediated conditional depletion of aga-miR-14 or aga-miR-305, but not aga-miR-8, increased mosquito resistance to both P. falciparum and P. berghei infection, and enhanced the mosquitoes' antibacterial defenses. Transcriptome analysis revealed that depletion of aga-miR-14 or aga-miR-305 resulted in an increased expression of multiple immunity-related and anti-Plasmodium genes in mosquito midguts. The overall fitness cost of conditionally expressed miR-SPs was low, with only one of eight fitness parameters being adversely affected. Taken together, our results demonstrate that targeting mosquito miRNA by conditional expression of miR-SPs may have potential for the development of malaria control through genetically engineered mosquitoes.


Subject(s)
Anopheles/immunology , Malaria, Falciparum/parasitology , MicroRNAs/immunology , Mosquito Vectors/immunology , Plasmodium berghei/physiology , Plasmodium falciparum/physiology , Animals , Anopheles/genetics , Anopheles/parasitology , Female , MicroRNAs/genetics , Mosquito Vectors/genetics , Mosquito Vectors/parasitology , Plasmodium berghei/genetics , Plasmodium berghei/immunology , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology
20.
Int Immunol ; 33(11): 587-594, 2021 10 29.
Article in English | MEDLINE | ID: mdl-34455438

ABSTRACT

Cerebral malaria (CM) is a life-threatening complication of the malaria disease caused by Plasmodium falciparum infection and is responsible for the death of half a million people annually. The molecular pathogenesis underlying CM in humans is not completely understood, although sequestration of infected erythrocytes in cerebral microvessels is thought to play a major role. In contrast, experimental cerebral malaria (ECM) models in mice have been thought to be distinct from human CM, and are mainly caused by inflammatory mediators. Here, to understand the spatial distribution and the potential sequestration of parasites in the whole-brain microvessels during a mouse model of ECM, we utilized the new tissue-clearing method CUBIC (Clear, Unobstructed, Brain/Body Imaging Cocktails and Computational analysis) with light-sheet fluorescent microscopy (LSFM), and reconstructed images in three dimensions (3D). We demonstrated significantly greater accumulation of Plasmodium berghei ANKA (PbANKA) parasites in the olfactory bulb (OB) of mice, compared with the other parts of the brain, including the cerebral cortex, cerebellum and brainstem. Furthermore, we show that PbANKA parasites preferentially accumulate in the brainstem when the OB is surgically removed. This study therefore not only highlights a successful application of CUBIC tissue-clearing technology to visualize the whole brain and its microvessels during ECM, but it also shows CUBIC's future potential for visualizing pathological events in the whole ECM brain at the cellular level, an achievement that would greatly advance our understanding of human cerebral malaria.


Subject(s)
Brain/pathology , Malaria, Cerebral/pathology , Animals , Brain/immunology , Brain/parasitology , Disease Models, Animal , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Plasmodium berghei/immunology
SELECTION OF CITATIONS
SEARCH DETAIL