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











Publication year range
1.
Comp Med ; 2023 Mar 07.
Article in English | MEDLINE | ID: mdl-36882188

ABSTRACT

C57BL/6J (B6) mice are commonly affected by ulcerative dermatitis (UD), a disease of unknown etiology with poor response to treatment. To study the possible role of diet in UD, we compared skin changes in B6 female mice fed a high-fat diet with those of mice fed a control diet. In addition, skin samples from mice with no, mild, moderate, and severe clinical signs of UD were examined by light and transmission electron microscopy (TEM). Mice fed a high-fat diet for 2 mo had more skin mast cell degranulation than did mice fed the control diet for the same period. Regardless of diet, older mice had more skin mast cells and more of these cells were degranulating as compared with younger mice. Microscopic changes in very early lesions were characterized by an increase in dermal mast cells and degranulation with focal areas of epidermal hyperplasia with or without hyperkeratosis. As the condition progressed, a mixed but predominantly neutrophilic inflammatory cell infiltrate appeared in the dermis, with or without epidermal erosion and scab formation. TEM showed that dermal mast cell membranes had disrupted and released of large number of electron dense granules, whereas degranulated mast cells were filled with isolated and coalescing empty spaces due to fusion of granule membranes. Ulceration appeared to occur very quickly, probably as result of intense scratching due to the pruritogenic properties of the histamine released from mast cell granules. This study showed a direct correlation between dietary fat and skin mast cell degranulation in female B6 mice. In addition, the number of skin mast cells and degranulation rates was higher in older mice. Treatments directed at preventing mast cell degranulation may result in better outcomes when applied early in UD cases. As noted previously in studies using caloric restriction, lower fat content in rodent diets may help prevent UD.

2.
mBio ; 14(2): e0339122, 2023 04 25.
Article in English | MEDLINE | ID: mdl-36852995

ABSTRACT

Cerebral malaria (CM), the deadliest complication of Plasmodium infection, is a complex and unpredictable disease. However, our understanding of the host and parasite factors that cause CM is limited. Using a mouse model of CM, experimental CM (ECM), we performed a three-way comparison between ECM-susceptible C57BL/6 mice infected with ECM-causing Plasmodium ANKA parasites [ANKA(C57BL/6)], ECM-resistant BALB/c mice infected with Plasmodium ANKA [ANKA(BALB/c)], and C57BL/6 mice infected with Plasmodium NK65 that does not cause ECM [NK65(C57BL/6)]. All ANKA(C57BL/6) mice developed CM. In contrast, in ANKA(BALB/c) and NK65(C57BL/6), infections do not result in CM and proceed similarly in terms of parasite growth, disease course, and host immune response. However, parasite gene expression in ANKA(BALB/c) was remarkably different than that in ANKA(C57BL/6) but similar to the gene expression in NK65(C57BL/6). Thus, Plasmodium ANKA has an ECM-specific gene expression profile that is activated only in susceptible hosts, providing evidence that the host has a critical influence on the outcome of infection. IMPORTANCE Hundreds of thousands of lives are lost each year due to the brain damage caused by malaria disease. The overwhelming majority of these deaths occur in young children living in sub-Saharan Africa. Thus far, there are no vaccines against this deadly disease, and we still do not know why fatal brain damage occurs in some children while others have milder, self-limiting disease progression. Our research provides an important clue to this problem. Here, we showed that the genetic background of the host has an important role in determining the course and the outcome of the disease. Our research also identified parasite molecules that can potentially be targeted in vaccination and therapy approaches.


Subject(s)
Malaria, Cerebral , Animals , Mice , Malaria, Cerebral/parasitology , Plasmodium berghei/physiology , Mice, Inbred C57BL , Gene Expression , Disease Models, Animal
3.
Sci Rep ; 10(1): 13630, 2020 08 12.
Article in English | MEDLINE | ID: mdl-32788672

ABSTRACT

Cerebral malaria (CM) is the deadliest form of severe Plasmodium infections. Currently, we have limited understanding of the mechanisms by which Plasmodium parasites induce CM. The mouse model of CM, experimental CM (ECM), induced by infection with the rodent parasite, Plasmodium berghei ANKA (PbANKA) has been extensively used to study the pathophysiology of CM. Recent genomic analyses revealed that the coding regions of PbANKA and the closely related Plasmodium berghei NK65 (PbNK65), that does not cause ECM, differ in only 21 single nucleotide polymorphysims (SNPs). Thus, the SNP-containing genes might contribute to the pathogenesis of ECM. Although the majority of these SNPs are located in genes of unknown function, one SNP is located in the DNA binding site of a member of the Plasmodium ApiAP2 transcription factor family, that we recently showed functions as a virulence factor alternating the host's immune response to the parasite. Here, we investigated the impact of this SNP on the development of ECM. Our results using CRISPR-Cas9 engineered parasites indicate that despite its immune modulatory function, the SNP is neither necessary nor sufficient to induce ECM and thus cannot account for parasite strain-specific differences in ECM phenotypes.


Subject(s)
CRISPR-Cas Systems/genetics , Extracellular Matrix/parasitology , Malaria, Cerebral/parasitology , Plasmodium berghei/genetics , Polymorphism, Single Nucleotide , Protozoan Proteins/genetics , Virulence Factors/genetics , Animals , Female , Mice , Mice, Inbred C57BL , Plasmodium berghei/growth & development , Plasmodium berghei/physiology , Protozoan Proteins/antagonists & inhibitors , Virulence Factors/antagonists & inhibitors
4.
Sci Adv ; 6(6): eaaw6957, 2020 02.
Article in English | MEDLINE | ID: mdl-32076635

ABSTRACT

The acquisition of malaria immunity is both remarkably slow and unpredictable. At present, we know little about the malaria parasite genes that influence the host's ability to mount a protective immune response. Here, we show that a single-nucleotide polymorphism (SNP) resulting in a single amino acid change (S to F) in an ApiAP2 transcription factor in the rodent malaria parasite Plasmodium berghei (Pb) NK65 allowed infected mice to mount a T helper cell 1 (TH1)-type immune response that controlled subsequent infections. As compared to PbNK65S, PbNK65F parasites differentially expressed 46 genes, most of which are predicted to play roles in immune evasion. PbNK65F infections resulted in an early interferon-γ response and a later expansion of germinal centers, resulting in high levels of infected red blood cell-specific TH1-type immunoglobulin G2b (IgG2b) and IgG2c antibodies. Thus, the Pb ApiAP2 transcription factor functions as a critical parasite virulence factor in malaria infections.


Subject(s)
Culicidae/parasitology , Host-Parasite Interactions/genetics , Host-Parasite Interactions/immunology , Immunity , Malaria/parasitology , Plasmodium berghei/genetics , Polymorphism, Single Nucleotide , Transcription Factor AP-2/genetics , Adaptive Immunity , Animals , DNA-Binding Proteins , Plasmodium berghei/metabolism , Protein Interaction Domains and Motifs , Th1 Cells/immunology , Th1 Cells/metabolism , Transcription Factor AP-2/chemistry , Transcription Factor AP-2/metabolism
5.
J Immunol ; 201(11): 3294-3306, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30373851

ABSTRACT

Activation of CD4+ T cells to proliferate drives cells toward aerobic glycolysis for energy production while using mitochondria primarily for macromolecular synthesis. In addition, the mitochondria of activated T cells increase production of reactive oxygen species, providing an important second messenger for intracellular signaling pathways. To better understand the critical changes in mitochondria that accompany prolonged T cell activation, we carried out an extensive analysis of mitochondrial remodeling using a combination of conventional strategies and a novel high-resolution imaging method. We show that for 4 d following activation, mouse CD4+ T cells sustained their commitment to glycolysis facilitated by increased glucose uptake through increased expression of GLUT transporters. Despite their limited contribution to energy production, mitochondria were active and showed increased reactive oxygen species production. Moreover, prolonged activation of CD4+ T cells led to increases in mitochondrial content and volume, in the number of mitochondria per cell and in mitochondrial biogenesis. Thus, during prolonged activation, CD4+ T cells continue to obtain energy predominantly from glycolysis but also undergo extensive mitochondrial remodeling, resulting in increased mitochondrial activity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Glutamate Plasma Membrane Transport Proteins/metabolism , Mitochondria/physiology , Reactive Oxygen Species/metabolism , Time Factors , Animals , Cells, Cultured , Energy Metabolism , Female , Glycolysis , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction
6.
Nat Immunol ; 19(8): 871-884, 2018 08.
Article in English | MEDLINE | ID: mdl-29988090

ABSTRACT

B cells are activated by two temporally distinct signals, the first provided by the binding of antigen to the B cell antigen receptor (BCR), and the second provided by helper T cells. Here we found that B cells responded to antigen by rapidly increasing their metabolic activity, including both oxidative phosphorylation and glycolysis. In the absence of a second signal, B cells progressively lost mitochondrial function and glycolytic capacity, which led to apoptosis. Mitochondrial dysfunction was a result of the gradual accumulation of intracellular calcium through calcium response-activated calcium channels that, for approximately 9 h after the binding of B cell antigens, was preventable by either helper T cells or signaling via the receptor TLR9. Thus, BCR signaling seems to activate a metabolic program that imposes a limited time frame during which B cells either receive a second signal and survive or are eliminated.


Subject(s)
B-Lymphocytes/physiology , Mitochondria/metabolism , Receptors, Antigen, B-Cell/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Toll-Like Receptor 9/metabolism , Animals , Apoptosis , Calcium/metabolism , Calcium Channels/metabolism , Cytokines/metabolism , Glycolysis , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NIH 3T3 Cells , Oxidative Phosphorylation , Receptors, Antigen, B-Cell/genetics , Signal Transduction , Toll-Like Receptor 9/genetics
7.
Nat Immunol ; 19(3): 255-266, 2018 03.
Article in English | MEDLINE | ID: mdl-29476183

ABSTRACT

Key events in T cell-dependent antibody responses, including affinity maturation, are dependent on the B cell's presentation of antigen to helper T cells at critical checkpoints in germinal-center formation in secondary lymphoid organs. Here we found that signaling via Toll-like receptor 9 (TLR9) blocked the ability of antigen-specific B cells to capture, process and present antigen and to activate antigen-specific helper T cells in vitro. In a mouse model in vivo and in a human clinical trial, the TLR9 agonist CpG enhanced the magnitude of the antibody response to a protein vaccine but failed to promote affinity maturation. Thus, TLR9 signaling might enhance antibody titers at the expense of the ability of B cells to engage in germinal-center events that are highly dependent on B cells' capture and presentation of antigen.


Subject(s)
Antibody Formation/immunology , Antigen Presentation/genetics , Lymphocyte Activation/immunology , Toll-Like Receptor 9/immunology , Animals , Antibody Affinity , Germinal Center/immunology , Humans , Malaria Vaccines , Mice , Toll-Like Receptor 9/agonists
8.
Eur J Immunol ; 47(11): 1890-1899, 2017 11.
Article in English | MEDLINE | ID: mdl-28762497

ABSTRACT

The development of vaccines for infectious diseases for which we currently have none, including HIV, will likely require the use of adjuvants that strongly promote germinal center responses and somatic hypermutation to produce broadly neutralizing antibodies. Here we compared the outcome of immunization with the T-cell dependent antigen, NP-conjugated to chicken gamma globulin (NP-CGG) adjuvanted with the toll-like receptor 9 (TLR9) ligands, CpG-A or CpG-B, alone or conjugated with the cationic lipid carrier, DOTAP. We provide evidence that only NP-CGG adjuvanted with DOTAP-CpG-B was an effective vaccine in mice resulting in robust germinal center responses, isotype switching and high affinity NP-specific antibodies. The effectiveness of DOTAP-CpG-B as an adjuvant was dependent on the expression of the TLR9 signaling adaptor MyD88 in immunized mice. These results indicate DOTAP-CpG-B but not DOTAP-CpG-A is an effective adjuvant for T cell-dependent protein antigen-based vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Fatty Acids, Monounsaturated/pharmacology , Oligodeoxyribonucleotides/immunology , Quaternary Ammonium Compounds/pharmacology , T-Lymphocytes/immunology , Vaccines/immunology , Animals , Antibody Affinity , Fatty Acids, Monounsaturated/immunology , Germinal Center/immunology , Mice , Mice, Inbred C57BL , Oligodeoxyribonucleotides/pharmacology , Quaternary Ammonium Compounds/immunology , Vaccines/pharmacology
9.
J Immunol ; 199(3): 931-940, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28652397

ABSTRACT

B cells express the innate receptor, TLR9, which signals in response to unmethylated CpG sequences in microbial DNA. Of the two major classes of CpG-containing oligonucleotides, CpG-A appears restricted to inducing type 1 IFN in innate immune cells and CpG-B to activating B cells to proliferate and produce Abs and inflammatory cytokines. Although CpGs are candidates for adjuvants to boost innate and adaptive immunity, our understanding of the effect of CpG-A and CpG-B on B cell responses is incomplete. In this study we show that both CpG-B and CpG-A activated B cells in vitro to proliferate, secrete Abs and IL-6, and that neither CpG-B nor CpG-A alone induced type 1 IFN production. However, when incorporated into the cationic lipid, DOTAP, CpG-A, but not CpG-B, induced a type 1 IFN response in B cells in vitro and in vivo. We provide evidence that differences in the function of CpG-A and CpG-B may be related to their intracellular trafficking in B cells. These findings fill an important gap in our understanding of the B cell response to CpGs, with implications for the use of CpG-A and CpG-B as immunomodulators.


Subject(s)
B-Lymphocytes/immunology , Interferon Type I/biosynthesis , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/immunology , Animals , Antibody Formation , B-Lymphocytes/drug effects , Cations/immunology , Cytokines/genetics , Cytokines/immunology , Immunity, Innate , Immunologic Factors/metabolism , Interferon Type I/immunology , Interleukin-6/biosynthesis , Interleukin-6/immunology , Lipids/administration & dosage , Lipids/chemistry , Lipids/pharmacology , Lymphocyte Activation , Mice , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/pharmacology , Toll-Like Receptor 9/agonists
10.
PLoS Pathog ; 12(12): e1006022, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27907215

ABSTRACT

Cerebral malaria (CM) is a severe complication of Plasmodium falciparum infection that results in thousands of deaths each year, mostly in African children. The in vivo mechanisms underlying this fatal condition are not entirely understood. Using the animal model of experimental cerebral malaria (ECM), we sought mechanistic insights into the pathogenesis of CM. Fatal disease was associated with alterations in tight junction proteins, vascular breakdown in the meninges / parenchyma, edema, and ultimately neuronal cell death in the brainstem, which is consistent with cerebral herniation as a cause of death. At the peak of ECM, we revealed using intravital two-photon microscopy that myelomonocytic cells and parasite-specific CD8+ T cells associated primarily with the luminal surface of CNS blood vessels. Myelomonocytic cells participated in the removal of parasitized red blood cells (pRBCs) from cerebral blood vessels, but were not required for the disease. Interestingly, the majority of disease-inducing parasite-specific CD8+ T cells interacted with the lumen of brain vascular endothelial cells (ECs), where they were observed surveying, dividing, and arresting in a cognate peptide-MHC I dependent manner. These activities were critically dependent on IFN-γ, which was responsible for activating cerebrovascular ECs to upregulate adhesion and antigen-presenting molecules. Importantly, parasite-specific CD8+ T cell interactions with cerebral vessels were impaired in chimeric mice rendered unable to present EC antigens on MHC I, and these mice were in turn resistant to fatal brainstem pathology. Moreover, anti-adhesion molecule (LFA-1 / VLA-4) therapy prevented fatal disease by rapidly displacing luminal CD8+ T cells from cerebrovascular ECs without affecting extravascular T cells. These in vivo data demonstrate that parasite-specific CD8+ T cell-induced fatal vascular breakdown and subsequent neuronal death during ECM is associated with luminal, antigen-dependent interactions with cerebrovasculature.


Subject(s)
Blood Vessels/pathology , Blood-Brain Barrier/pathology , Brain Stem/pathology , CD8-Positive T-Lymphocytes/pathology , Malaria, Cerebral/pathology , Animals , Disease Models, Animal , Flow Cytometry , Immunohistochemistry , Malaria, Cerebral/immunology , Mice , Mice, Transgenic , Plasmodium berghei
11.
Immunity ; 44(6): 1365-78, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27261276

ABSTRACT

Receptor CD300b is implicated in regulating the immune response to bacterial infection by an unknown mechanism. Here, we identified CD300b as a lipopolysaccharide (LPS)-binding receptor and determined the mechanism underlying CD300b augmentation of septic shock. In vivo depletion and adoptive transfer studies identified CD300b-expressing macrophages as the key cell type augmenting sepsis. We showed that CD300b, and its adaptor DAP12, associated with Toll-like receptor 4 (TLR4) upon LPS binding, thereby enhancing TLR4-adaptor MyD88- and TRIF-dependent signaling that resulted in an elevated pro-inflammatory cytokine storm. LPS engagement of the CD300b-TLR4 complex led to the recruitment and activation of spleen tyrosine kinase (Syk) and phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K). This resulted in an inhibition of the ERK1/2 protein kinase- and NF-κB transcription factor-mediated signaling pathways, which subsequently led to a reduced interleukin-10 (IL-10) production. Collectively, our data describe a mechanism of TLR4 signaling regulated by CD300b in myeloid cells in response to LPS.


Subject(s)
Interleukin-10/metabolism , Macrophages/immunology , Peritonitis/immunology , Receptors, Immunologic/metabolism , Sepsis/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , HEK293 Cells , Humans , Interleukin-10/genetics , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Receptors, Immunologic/genetics , Signal Transduction , Syk Kinase/metabolism , Toll-Like Receptor 4/metabolism
12.
Proc Natl Acad Sci U S A ; 112(42): 13075-80, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26438846

ABSTRACT

The most deadly complication of Plasmodium falciparum infection is cerebral malaria (CM) with a case fatality rate of 15-25% in African children despite effective antimalarial chemotherapy. There are no adjunctive treatments for CM, so there is an urgent need to identify new targets for therapy. Here we show that the glutamine analog 6-diazo-5-oxo-L-norleucine (DON) rescues mice from CM when administered late in the infection a time at which mice already are suffering blood-brain barrier dysfunction, brain swelling, and hemorrhaging accompanied by accumulation of parasite-specific CD8(+) effector T cells and infected red blood cells in the brain. Remarkably, within hours of DON treatment mice showed blood-brain barrier integrity, reduced brain swelling, decreased function of activated effector CD8(+) T cells in the brain, and levels of brain metabolites that resembled those in uninfected mice. These results suggest DON as a strong candidate for an effective adjunctive therapy for CM in African children.


Subject(s)
Antimalarials/therapeutic use , Diazooxonorleucine/therapeutic use , Glutamine/metabolism , Malaria, Cerebral/drug therapy , Malaria, Falciparum/drug therapy , Animals , Antimalarials/pharmacology , Blood-Brain Barrier/drug effects , Diazooxonorleucine/pharmacology , Malaria, Cerebral/metabolism , Malaria, Falciparum/metabolism , Mice
13.
mBio ; 6(3): e00725, 2015 Jun 02.
Article in English | MEDLINE | ID: mdl-26037126

ABSTRACT

UNLABELLED: Malaria is an infectious disease caused by parasites of several Plasmodium spp. Cerebral malaria (CM) is a common form of severe malaria resulting in nearly 700,000 deaths each year in Africa alone. At present, there is no adjunctive therapy for CM. Although the mechanisms underlying the pathogenesis of CM are incompletely understood, it is likely that both intrinsic features of the parasite and the human host's immune response contribute to disease. The kinase mammalian target of rapamycin (mTOR) is a central regulator of immune responses, and drugs that inhibit the mTOR pathway have been shown to be antiparasitic. In a mouse model of CM, experimental CM (ECM), we show that the mTOR inhibitor rapamycin protects against ECM when administered within the first 4 days of infection. Treatment with rapamycin increased survival, blocked breakdown of the blood-brain barrier and brain hemorrhaging, decreased the influx of both CD4(+) and CD8(+) T cells into the brain and the accumulation of parasitized red blood cells in the brain. Rapamycin induced marked transcriptional changes in the brains of infected mice, and analysis of transcription profiles predicted that rapamycin blocked leukocyte trafficking to and proliferation in the brain. Remarkably, animals were protected against ECM even though rapamycin treatment significantly increased the inflammatory response induced by infection in both the brain and spleen. These results open a new avenue for the development of highly selective adjunctive therapies for CM by targeting pathways that regulate host and parasite metabolism. IMPORTANCE: Malaria is a highly prevalent infectious disease caused by parasites of several Plasmodium spp. Malaria is usually uncomplicated and resolves with time; however, in about 1% of cases, almost exclusively among young children, malaria becomes severe and life threatening, resulting in nearly 700,000 deaths each year in Africa alone. Among the most severe complications of Plasmodium falciparum infection is cerebral malaria with a fatality rate of 15 to 20%, despite treatment with antimalarial drugs. Cerebral malaria takes a second toll on African children, leaving survivors at high risk of debilitating neurological defects. At present, we have no effective adjunctive therapies for cerebral malaria, and developing such therapies would have a large impact on saving young lives in Africa. Here we report results that open a new avenue for the development of highly selective adjunctive therapies for cerebral malaria by targeting pathways that regulate host and parasite metabolism.


Subject(s)
Malaria, Cerebral/prevention & control , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Brain/pathology , Gene Expression Profiling , Malaria, Cerebral/pathology , Mice , Survival Analysis
14.
PLoS One ; 9(2): e87140, 2014.
Article in English | MEDLINE | ID: mdl-24586264

ABSTRACT

BACKGROUND: The role of intracellular radical oxygen species (ROS) in pathogenesis of cerebral malaria (CM) remains incompletely understood. METHODS AND FINDINGS: We undertook testing Tempol--a superoxide dismutase (SOD) mimetic and pleiotropic intracellular antioxidant--in cells relevant to malaria pathogenesis in the context of coagulation and inflammation. Tempol was also tested in a murine model of CM induced by Plasmodium berghei Anka infection. Tempol was found to prevent transcription and functional expression of procoagulant tissue factor in endothelial cells (ECs) stimulated by lipopolysaccharide (LPS). This effect was accompanied by inhibition of IL-6, IL-8, and monocyte chemoattractant protein (MCP-1) production. Tempol also attenuated platelet aggregation and human promyelocytic leukemia HL60 cells oxidative burst. In dendritic cells, Tempol inhibited LPS-induced production of TNF-α, IL-6, and IL-12p70, downregulated expression of co-stimulatory molecules, and prevented antigen-dependent lymphocyte proliferation. Notably, Tempol (20 mg/kg) partially increased the survival of mice with CM. Mechanistically, treated mice had lowered plasma levels of MCP-1, suggesting that Tempol downmodulates EC function and vascular inflammation. Tempol also diminished blood brain barrier permeability associated with CM when started at day 4 post infection but not at day 1, suggesting that ROS production is tightly regulated. Other antioxidants-such as α-phenyl N-tertiary-butyl nitrone (PBN; a spin trap), MnTe-2-PyP and MnTBAP (Mn-phorphyrin), Mitoquinone (MitoQ) and Mitotempo (mitochondrial antioxidants), M30 (an iron chelator), and epigallocatechin gallate (EGCG; polyphenol from green tea) did not improve survival. By contrast, these compounds (except PBN) inhibited Plasmodium falciparum growth in culture with different IC50s. Knockout mice for SOD1 or phagocyte nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (gp91(phox-/-)) or mice treated with inhibitors of SOD (diethyldithiocarbamate) or NADPH oxidase (diphenyleneiodonium) did not show protection or exacerbation for CM. CONCLUSION: Results with Tempol suggest that intracellular ROS contribute, in part, to CM pathogenesis. Therapeutic targeting of intracellular ROS in CM is discussed.


Subject(s)
Antioxidants/pharmacology , Cyclic N-Oxides/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Malaria, Cerebral/drug therapy , Thromboplastin/metabolism , Animals , Antioxidants/therapeutic use , Cells, Cultured , Chemokine CCL2/metabolism , Cyclic N-Oxides/therapeutic use , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Malaria, Cerebral/metabolism , Mice , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Spin Labels
15.
PLoS One ; 8(5): e62820, 2013.
Article in English | MEDLINE | ID: mdl-23675429

ABSTRACT

Severe malaria, including cerebral malaria (CM) and placental malaria (PM), have been recognized to have many of the features of uncontrolled inflammation. We recently showed that in mice genetic susceptibility to the lethal inflammatory autoimmune disease, systemic lupus erythematosus (SLE), conferred resistance to CM. Protection appeared to be mediated by immune mechanisms that allowed SLE-prone mice, prior to the onset of overt SLE symptoms, to better control their inflammatory response to Plasmodium infection. Here we extend these findings to ask does SLE susceptibility have 1) a cost to reproductive fitness and/or 2) an effect on PM in mice? The rates of conception for WT and SLE susceptible (SLE(s)) mice were similar as were the number and viability of fetuses in pregnant WT and SLE(s) mice indicating that SLE susceptibility does not have a reproductive cost. We found that Plasmodium chabaudi AS (Pc) infection disrupted early stages of pregnancy before the placenta was completely formed resulting in massive decidual necrosis 8 days after conception. Pc-infected pregnant SLE(s) mice had significantly more fetuses (∼1.8 fold) but SLE did not significantly affect fetal viability in infected animals. This was despite the fact that Pc-infected pregnant SLE(s) mice had more severe symptoms of malaria as compared to Pc-infected pregnant WT mice. Thus, although SLE susceptibility was not protective in PM in mice it also did not have a negative impact on reproductive fitness.


Subject(s)
Genetic Predisposition to Disease , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Malaria/genetics , Malaria/immunology , Placenta/parasitology , Pregnancy Complications, Parasitic , Animals , Disease Models, Animal , Disease Susceptibility , Erythrocyte Indices , Female , Lupus Erythematosus, Systemic/complications , Malaria/blood , Malaria/complications , Mice , Parasitemia/genetics , Parasitemia/immunology , Placenta/pathology , Pregnancy , Reproduction/genetics , Reproduction/immunology , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL