Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(1)2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36614196

ABSTRACT

Large numbers of neutrophils infiltrate tumors and comprise a notable component of the inflammatory tumor microenvironment. While it is established that tumor cells exhibit the Warburg effect for energy production, the contribution of the neutrophil metabolic state to tumorigenesis is unknown. Here, we investigated whether neutrophil infiltration and metabolic status promotes tumor progression in an orthotopic mouse model of pancreatic ductal adenocarcinoma (PDAC). We observed a large increase in the proportion of neutrophils in the blood and tumor upon orthotopic transplantation. Intriguingly, these tumor-infiltrating neutrophils up-regulated glycolytic factors and hypoxia-inducible factor 1-alpha (HIF-1α) expression compared to neutrophils from the bone marrow and blood of the same mouse. This enhanced glycolytic signature was also observed in human PDAC tissue samples. Strikingly, neutrophil-specific deletion of HIF-1α (HIF-1αΔNφ) significantly reduced tumor burden and improved overall survival in orthotopic transplanted mice, by converting the pro-tumorigenic neutrophil phenotype to an anti-tumorigenic phenotype. This outcome was associated with elevated reactive oxygen species production and activated natural killer cells and CD8+ cytotoxic T cells compared to littermate control mice. These data suggest a role for HIF-1α in neutrophil metabolism, which could be exploited as a target for metabolic modulation in cancer.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Animals , Mice , Neutrophils/metabolism , Cell Line, Tumor , Mice, Knockout , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Carcinogenesis , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Tumor Microenvironment/genetics , Pancreatic Neoplasms
2.
Int J Mol Sci ; 22(12)2021 Jun 14.
Article in English | MEDLINE | ID: mdl-34198548

ABSTRACT

Inflammation in the tumor microenvironment has been shown to promote disease progression in pancreatic ductal adenocarcinoma (PDAC); however, the role of macrophage metabolism in promoting inflammation is unclear. Using an orthotopic mouse model of PDAC, we demonstrate that macrophages from tumor-bearing mice exhibit elevated glycolysis. Macrophage-specific deletion of Glucose Transporter 1 (GLUT1) significantly reduced tumor burden, which was accompanied by increased Natural Killer and CD8+ T cell activity and suppression of the NLRP3-IL1ß inflammasome axis. Administration of mice with a GLUT1-specific inhibitor reduced tumor burden, comparable with gemcitabine, the current standard-of-care. In addition, we observe that intra-tumoral macrophages from human PDAC patients exhibit a pronounced glycolytic signature, which reliably predicts poor survival. Our data support a key role for macrophage metabolism in tumor immunity, which could be exploited to improve patient outcomes.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , Cytoprotection , Glycolysis , Macrophages/metabolism , Pancreatic Neoplasms/pathology , Adenocarcinoma/immunology , Animals , Carcinoma, Pancreatic Ductal/immunology , Cell Proliferation/drug effects , Cytoprotection/drug effects , Drug Resistance, Neoplasm/drug effects , Glucose Transporter Type 1/metabolism , Glycolysis/drug effects , Humans , Hydroxybenzoates/pharmacology , Inflammation/pathology , Interleukin-1beta/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Macrophages/drug effects , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pancreatic Neoplasms/immunology , Survival Analysis , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Tumor Burden/drug effects , Pancreatic Neoplasms
3.
Redox Biol ; 25: 101174, 2019 07.
Article in English | MEDLINE | ID: mdl-30917934

ABSTRACT

Recent years have seen a renaissance in the research linking inflammation and cancer with immune cells playing a central role in smouldering inflammation in the tumor microenvironment. Diverse immune cell types infiltrate the tumor microenvironment, and the dynamic tumor-immune cell interplay gives rise to a rich milieu of cytokines and growth factors. Fundamentally, this intricate cross-talk creates the conducive condition for tumor cell proliferation, survival and metastasis. Interestingly, the prominent impact of immune cells is expounded in their contrary pro-tumoral role, as well as their potential anti-cancer cellular weaponry. The latter is known as immunotherapy, a concept born out of evidence that tumors are susceptible to immune defence and that by manipulating the immune system, tumor growth can be successfully restrained. Naturally, a deeper understanding of the multifaceted roles of various immune cell types thus contributes toward developing innovative anti-cancer strategies. Therefore, in this review we first outline the roles played by the major immune cell types, such as macrophages, neutrophils, natural killer cells, T cells and B cells. We then explain the recently-explored strategies of immunomodulation and discuss some important approaches via an immunology perspective.


Subject(s)
Immunotherapy , Lymphocytes/immunology , Neoplasms/immunology , Neoplasms/therapy , Animals , Cell Cycle Checkpoints , Humans , Nanoparticles/chemistry
4.
Front Cell Dev Biol ; 6: 168, 2018.
Article in English | MEDLINE | ID: mdl-30619850

ABSTRACT

Immune cells are one of the most versatile cell types, as they can tailor their metabolic activity according to their required function. In response to diverse environmental cues, immune cells undergo metabolic reprogramming to support their differentiation, proliferation and pro-inflammatory effector functions. To meet a dramatic surge in energetic demand, immune cells rewire their metabolism to utilize aerobic glycolysis. This preferential use of glycolysis even under aerobic conditions is well established in tumor cells, and is known as the "Warburg effect." Tumor cells avidly use glucose for aerobic glycolysis, thereby creating a nutrient-starved microenvironment, outcompeting T cells for glucose, and directly inhibiting T-cell anti-tumoral effector function. Given that both immune and tumor cells use similar modes of metabolism in the tumor stroma, it is imperative to identify a therapeutic window in which immune-cell and tumor-cell glycolysis can be specifically targeted. In this review, we focus on the Warburg metabolism as well as other metabolic pathways of myeloid cells, which comprise a notable niche in the tumor environment and promote the growth and metastasis of malignant tumors. We examine how differential immune-cell activation triggers metabolic fate, and detail how this forbidding microenvironment succeeds in shutting down the vigorous anti-tumoral response. Finally, we highlight emerging therapeutic concepts that aim to target immune-cell metabolism. Improving our understanding of immunometabolism and immune-cell commitment to specific metabolic fates will help identify alternative therapeutic approaches to battle this intractable disease.

5.
EMBO Mol Med ; 10(1): 121-138, 2018 01.
Article in English | MEDLINE | ID: mdl-29113976

ABSTRACT

Arboviral diseases have risen significantly over the last 40 years, increasing the risk of co-infection with other endemic disease such as malaria. However, nothing is known about the impact arboviruses have on the host response toward heterologous pathogens during co-infection. Here, we investigate the effects of Chikungunya virus (CHIKV) co-infection on the susceptibility and severity of malaria infection. Using the Plasmodium berghei ANKA (PbA) experimental cerebral malaria (ECM) model, we show that concurrent co-infection induced the most prominent changes in ECM manifestation. Concurrent co-infection protected mice from ECM mortality without affecting parasite development in the blood. This protection was mediated by the alteration of parasite-specific CD8+ T-cell trafficking through an IFNγ-mediated mechanism. Co-infection with CHIKV induced higher splenic IFNγ levels that lead to high local levels of CXCL9 and CXCL10. This induced retention of CXCR3-expressing pathogenic CD8+ T cells in the spleen and prevented their migration to the brain. This then averts all downstream pathogenic events such as parasite sequestration in the brain and disruption of blood-brain barrier that prevents ECM-induced mortality in co-infected mice.


Subject(s)
Brain/pathology , CD8-Positive T-Lymphocytes/pathology , Chikungunya Fever/pathology , Chikungunya virus/physiology , Coinfection/pathology , Malaria, Cerebral/pathology , Plasmodium berghei/physiology , Animals , Brain/parasitology , Brain/virology , CD8-Positive T-Lymphocytes/parasitology , CD8-Positive T-Lymphocytes/virology , Cell Movement , Chikungunya Fever/parasitology , Chikungunya Fever/virology , Coinfection/parasitology , Coinfection/virology , Female , Malaria, Cerebral/parasitology , Malaria, Cerebral/virology , Male , Mice , Mice, Inbred C57BL , Neuropathology , Protective Factors
6.
Cell Microbiol ; 20(5): e12819, 2018 05.
Article in English | MEDLINE | ID: mdl-29281764

ABSTRACT

Host immune response has a key role in controlling the progression of malaria infection. In the well-established murine model of experimental cerebral malaria (ECM) with Plasmodium berghei ANKA infection, proinflammatory Th1 and CD8+ T cell response are essential for disease development. Interferon regulatory factor 1 (IRF1) is a transcription factor that promotes Th1 responses, and its absence was previously shown to protect from ECM death. Yet the exact mechanism of protection remains unknown. Here we demonstrated that IRF1-deficient mice (IRF1 knockout) were protected from ECM death despite displaying early neurological signs. Resistance to ECM death was a result of reduced parasite sequestration and pathogenic CD8+ T cells in the brain. Further analysis revealed that IRF1 deficiency suppress interferon-γ production and delayed CD8+ T cell proliferation. CXCR3 expression was found to be decreased in pathogenic CD8+ T cells, which limited their migration to the brain. In addition, reduced expression of adhesion molecules by brain endothelial cells hampered leucocyte retention in the brain. Taken together, these factors limited sequestration of pathogenic CD8+ T cells and consequently its ability to induce extensive damage to the blood-brain barrier.


Subject(s)
Interferon Regulatory Factor-1/genetics , Malaria, Cerebral/genetics , Plasmodium berghei/pathogenicity , Receptors, CXCR3/genetics , Animals , Brain/microbiology , Brain/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/microbiology , Cell Movement/genetics , Disease Models, Animal , Gene Expression Regulation , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Malaria, Cerebral/immunology , Malaria, Cerebral/microbiology , Mice , Mice, Knockout
7.
Exp Mol Med ; 49(2): e292, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28186086

ABSTRACT

Given that increased thermogenesis in white adipose tissue, also known as browning, promotes energy expenditure, significant efforts have been invested to determine the molecular factors involved in this process. Here we show that HOXC10, a homeobox domain-containing transcription factor expressed in subcutaneous white adipose tissue, is a suppressor of genes involved in browning white adipose tissue. Ectopic expression of HOXC10 in adipocytes suppresses brown fat genes, whereas the depletion of HOXC10 in adipocytes and myoblasts increases the expression of brown fat genes. The protein level of HOXC10 inversely correlates with brown fat genes in subcutaneous white adipose tissue of cold-exposed mice. Expression of HOXC10 in mice suppresses cold-induced browning in subcutaneous white adipose tissue and abolishes the beneficial effect of cold exposure on glucose clearance. HOXC10 exerts its effect, at least in part, by suppressing PRDM16 expression. The results support that HOXC10 is a key negative regulator of the process of browning in white adipose tissue.


Subject(s)
Adipose Tissue, White/physiology , Gene Expression Regulation , Homeodomain Proteins/metabolism , Thermogenesis , Adipocytes/metabolism , Adipose Tissue, Brown/physiology , Animals , Cell Line , Cold Temperature , Energy Metabolism , Glucose/metabolism , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Male , Mice , Mice, Inbred C57BL
8.
Nat Protoc ; 10(12): 2016-26, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26562622

ABSTRACT

We have recently demonstrated that brain endothelial cells cross-present parasite antigen during mouse experimental cerebral malaria (ECM). Here we describe a 2-d protocol to detect cross-presentation by isolating the brain microvessels and incubating them with a reporter cell line that expresses lacZ upon detection of the relevant peptide-major histocompatibility complex. After X-gal staining, a typical positive result consists of hundreds of blue spots, compared with fewer than 20 spots from a naive brain. The assay is generalizable to other disease contexts by using reporter cells that express appropriate specific T cell receptors. Also described is the protocol for culturing endothelial cells from brain microvessels isolated from naive mice. After 7-10 d, an in vitro cross-presentation assay can be performed by adding interferon-γ, antigen (e.g., Plasmodium berghei-infected red blood cells) and reporter cells in sequence over 3 d. This is useful for comparing different antigen forms or for probing the effects of various interventions.


Subject(s)
Antigen Presentation , Brain/immunology , Endothelial Cells/immunology , Microvessels/immunology , Animals , Brain/blood supply , Brain/cytology , Cell Culture Techniques , Cell Line , Cells, Cultured , Disease Models, Animal , Endothelial Cells/cytology , Major Histocompatibility Complex , Malaria/immunology , Mice , Mice, Inbred C57BL , Microvessels/cytology , Plasmodium berghei/immunology , Receptors, Antigen, T-Cell/immunology
9.
Semin Immunopathol ; 37(3): 221-31, 2015 May.
Article in English | MEDLINE | ID: mdl-25772948

ABSTRACT

Cerebral malaria (CM) is one the major complications occurring during malaria infection. The mechanisms leading to this syndrome are still not completely understood. Although it is clear that parasite sequestration is the key initiation factor, the downstream pathological processes are still highly debated. The experimental cerebral malaria (ECM) model, in which susceptible mice are infected with Plasmodium berghei ANKA, has led to the identification of CD8(+) T cells as the major mediator of ECM death. In this review, we discuss the recent advances and future developments in the understanding of the role of CD8(+) T cells in CM.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Host-Parasite Interactions/immunology , Malaria, Cerebral/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Movement , Cytotoxicity, Immunologic , Disease Models, Animal , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Extracellular Matrix/immunology , Humans , Immunomodulation , Malaria, Cerebral/parasitology , Phenotype , Plasmodium/immunology
10.
Eur J Immunol ; 45(1): 130-41, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25319247

ABSTRACT

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


Subject(s)
Brain/immunology , Interferon Regulatory Factor-7/immunology , Malaria, Cerebral/immunology , Receptor, Interferon alpha-beta/immunology , Spleen/immunology , Th1 Cells/immunology , Animals , Antibodies, Monoclonal/pharmacology , Brain/drug effects , Brain/parasitology , Disease Susceptibility , Erythrocytes/parasitology , Female , Gene Expression Regulation , Host-Parasite Interactions , Interferon Regulatory Factor-7/genetics , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Plasmodium berghei/immunology , Plasmodium chabaudi/immunology , Receptor, Interferon alpha-beta/antagonists & inhibitors , Receptor, Interferon alpha-beta/genetics , Signal Transduction , Spleen/drug effects , Spleen/parasitology , Th1 Cells/parasitology , Time Factors
11.
Mediators Inflamm ; 2014: 243713, 2014.
Article in English | MEDLINE | ID: mdl-25157202

ABSTRACT

Malaria is one of the most serious infectious diseases in humans and responsible for approximately 500 million clinical cases and 500 thousand deaths annually. Acquired adaptive immune responses control parasite replication and infection-induced pathologies. Most infections are clinically silent which reflects on the ability of adaptive immune mechanisms to prevent the disease. However, a minority of these can become severe and life-threatening, manifesting a range of overlapping syndromes of complex origins which could be induced by uncontrolled immune responses. Major players of the innate and adaptive responses are interferons. Here, we review their roles and the signaling pathways involved in their production and protection against infection and induced immunopathologies.


Subject(s)
Interferon Regulatory Factors/metabolism , Interferons/metabolism , Malaria/metabolism , Animals , Humans
12.
J Clin Invest ; 124(6): 2483-96, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24789914

ABSTRACT

Many pathogens, including viruses, bacteria, and protozoan parasites, suppress cellular immune responses through activation of type I IFN signaling. Recent evidence suggests that immune suppression and susceptibility to the malaria parasite, Plasmodium, is mediated by type I IFN; however, it is unclear how type I IFN suppresses immunity to blood-stage Plasmodium parasites. During experimental severe malaria, CD4+ Th cell responses are suppressed, and conventional DC (cDC) function is curtailed through unknown mechanisms. Here, we tested the hypothesis that type I IFN signaling directly impairs cDC function during Plasmodium infection in mice. Using cDC-specific IFNAR1-deficient mice, and mixed BM chimeras, we found that type I IFN signaling directly affects cDC function, limiting the ability of cDCs to prime IFN-γ-producing Th1 cells. Although type I IFN signaling modulated all subsets of splenic cDCs, CD8- cDCs were especially susceptible, exhibiting reduced phagocytic and Th1-promoting properties in response to type I IFNs. Additionally, rapid and systemic IFN-α production in response to Plasmodium infection required type I IFN signaling in cDCs themselves, revealing their contribution to a feed-forward cytokine-signaling loop. Together, these data suggest abrogation of type I IFN signaling in CD8- splenic cDCs as an approach for enhancing Th1 responses against Plasmodium and other type I IFN-inducing pathogens.


Subject(s)
Dendritic Cells/immunology , Interferon Type I/metabolism , Malaria/immunology , Th1 Cells/immunology , Animals , CD8 Antigens/metabolism , Dendritic Cells/classification , Female , Immune Tolerance , Immunity, Cellular , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Plasmodium berghei/immunology , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Signal Transduction/immunology
13.
Parasitol Int ; 63(1): 187-94, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23892178

ABSTRACT

Malaria, a disease caused by the Plasmodium parasite, remains one of the most deadly infectious diseases known to mankind. The parasite has a complex life cycle, of which only the erythrocytic stage is responsible for the diverse pathologies induced during infection. To date, the disease mechanisms that underlie these pathologies are still poorly understood. In the case of infections caused by Plasmodium falciparum, the species responsible for most malaria related deaths, pathogenesis is thought to be due to the sequestration of infected red blood cells (IRBCs) in deep tissues. Other human and rodent malaria parasite species are also known to exhibit sequestration. Here, we review the different techniques that allow researchers to study how rodent malaria parasites modify their host cells, the distribution of IRBCs in vivo as well as the interactions between IRBCs and host tissues.


Subject(s)
Luminescent Measurements/methods , Magnetic Resonance Imaging/methods , Microscopy/methods , Plasmodium/cytology , Positron-Emission Tomography/methods , Animals , Plasmodium/physiology , Rodentia
14.
EMBO Mol Med ; 5(7): 984-99, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23681698

ABSTRACT

Cerebral malaria is a devastating complication of Plasmodium falciparum infection. Its pathogenesis is complex, involving both parasite- and immune-mediated events. CD8(+) T cells play an effector role in murine experimental cerebral malaria (ECM) induced by Plasmodium berghei ANKA (PbA) infection. We have identified a highly immunogenic CD8 epitope in glideosome-associated protein 50 that is conserved across rodent malaria species. Epitope-specific CD8(+) T cells are induced during PbA infection, migrating to the brain just before neurological signs manifest. They are functional, cytotoxic and can damage the blood-brain barrier in vivo. Such CD8(+) T cells are also found in the brain during infection with parasite strains/species that do not induce neuropathology. We demonstrate here that PbA infection causes brain microvessels to cross-present parasite antigen, while non-ECM-causing parasites do not. Further, treatment with fast-acting anti-malarial drugs before the onset of ECM reduces parasite load and thus antigen presentation in the brain, preventing ECM death. Thus our data suggest that combined therapies targeting both the parasite and host antigen-presenting cells may improve the outcome of CM patients.


Subject(s)
Antigens, Protozoan/immunology , Brain/blood supply , Brain/parasitology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Amino Acid Sequence , Animals , Antigens, Protozoan/analysis , Antimalarials/therapeutic use , Blood-Brain Barrier/immunology , Blood-Brain Barrier/parasitology , Blood-Brain Barrier/pathology , Brain/immunology , Brain/pathology , CD8-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/parasitology , Cross-Priming/drug effects , Female , Humans , Malaria, Cerebral/drug therapy , Malaria, Cerebral/parasitology , Malaria, Cerebral/pathology , Mice , Mice, Inbred C57BL , Microvessels/immunology , Microvessels/parasitology , Microvessels/pathology , Molecular Sequence Data , Parasite Load , Plasmodium berghei/drug effects , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Receptors, Antigen, T-Cell, alpha-beta/immunology
15.
PLoS One ; 6(4): e18720, 2011 Apr 11.
Article in English | MEDLINE | ID: mdl-21494565

ABSTRACT

BACKGROUND: Infection with Plasmodium berghei ANKA (PbA) in susceptible mice induces a syndrome called experimental cerebral malaria (ECM) with severe pathologies occurring in various mouse organs. Immune mediators such as T cells or cytokines have been implicated in the pathogenesis of ECM. Red blood cells infected with PbA parasites have been shown to accumulate in the brain and other tissues during infection. This accumulation is thought to be involved in PbA-induced pathologies, which mechanisms are poorly understood. METHODS AND FINDINGS: Using transgenic PbA parasites expressing the luciferase protein, we have assessed by real-time in vivo imaging the dynamic and temporal contribution of different immune factors in infected red blood cell (IRBC) accumulation and distribution in different organs during PbA infection. Using deficient mice or depleting antibodies, we observed that CD8(+) T cells and IFN-γ drive the rapid increase in total parasite biomass and accumulation of IRBC in the brain and in different organs 6-12 days post-infection, at a time when mice develop ECM. Other cells types like CD4(+) T cells, monocytes or neutrophils or cytokines such as IL-12 and TNF-α did not influence the early increase of total parasite biomass and IRBC accumulation in different organs. CONCLUSIONS: CD8(+) T cells and IFN-γ are the major immune mediators controlling the time-dependent accumulation of P. berghei-infected red blood cells in tissues.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Erythrocytes/parasitology , Interferon-gamma/metabolism , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Organ Specificity/immunology , Animals , Biomass , Brain/immunology , Brain/parasitology , Brain/pathology , DNA-Binding Proteins/metabolism , Erythrocytes/immunology , Female , Lymphocyte Depletion , Male , Mice , Mice, Inbred C57BL , Myeloid Cells/immunology , Plasmodium berghei/physiology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...