Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Immunity ; 56(3): 592-605.e8, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36804959

ABSTRACT

Plasmodium replicates within the liver prior to reaching the bloodstream and infecting red blood cells. Because clinical manifestations of malaria only arise during the blood stage of infection, a perception exists that liver infection does not impact disease pathology. By developing a murine model where the liver and blood stages of infection are uncoupled, we showed that the integration of signals from both stages dictated mortality outcomes. This dichotomy relied on liver stage-dependent activation of Vγ4+ γδ T cells. Subsequent blood stage parasite loads dictated their cytokine profiles, where low parasite loads preferentially expanded IL-17-producing γδ T cells. IL-17 drove extra-medullary erythropoiesis and concomitant reticulocytosis, which protected mice from lethal experimental cerebral malaria (ECM). Adoptive transfer of erythroid precursors could rescue mice from ECM. Modeling of γδ T cell dynamics suggests that this protective mechanism may be key for the establishment of naturally acquired malaria immunity among frequently exposed individuals.


Subject(s)
Erythropoiesis , Malaria, Cerebral , Animals , Mice , Erythrocytes , Interleukin-17 , Liver/parasitology , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, gamma-delta , Malaria
2.
Elife ; 112022 12 07.
Article in English | MEDLINE | ID: mdl-36476511

ABSTRACT

Anthracyclines are among the most used and effective anticancer drugs. Their activity has been attributed to DNA double-strand breaks resulting from topoisomerase II poisoning and to eviction of histones from select sites in the genome. Here, we show that the extensively used anthracyclines Doxorubicin, Daunorubicin, and Epirubicin decrease the transcription of nuclear factor kappa B (NF-κB)-dependent gene targets, but not interferon-responsive genes in primary mouse (Mus musculus) macrophages. Using an NMR-based structural approach, we demonstrate that anthracyclines disturb the complexes formed between the NF-κB subunit RelA and its DNA-binding sites. The anthracycline variants Aclarubicin, Doxorubicinone, and the newly developed Dimethyl-doxorubicin, which share anticancer properties with the other anthracyclines but do not induce DNA damage, also suppressed inflammation, thus uncoupling DNA damage from the effects on inflammation. These findings have implications for anticancer therapy and for the development of novel anti-inflammatory drugs with limited side effects for life-threatening conditions such as sepsis.


Subject(s)
Anthracyclines , NF-kappa B , Animals , Mice , Anthracyclines/pharmacology , Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , DNA Damage , DNA
3.
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
4.
Nat Commun ; 13(1): 3747, 2022 06 29.
Article in English | MEDLINE | ID: mdl-35768411

ABSTRACT

Severe malaria can manifest itself with a variety of well-recognized clinical phenotypes that are highly predictive of death - severe anaemia, coma (cerebral malaria), multiple organ failure, and respiratory distress. The reasons why an infected individual develops one pathology rather than another remain poorly understood. Here we use distinct rodent models of infection to show that the host microbiota is a contributing factor for the development of respiratory distress syndrome and host mortality in the context of malaria infections (malaria-associated acute respiratory distress syndrome, MA-ARDS). We show that parasite sequestration in the lung results in sustained immune activation. Subsequent production of the anti-inflammatory cytokine IL-10 by T cells compromises microbial control, leading to severe lung disease. Notably, bacterial clearance with linezolid, an antibiotic commonly used in the clinical setting to control lung-associated bacterial infections, prevents MA-ARDS-associated lethality. Thus, we propose that the host's anti-inflammatory response to limit tissue damage can result in loss of microbial control, which promotes MA-ARDS. This must be considered when intervening against life-threatening respiratory complications.


Subject(s)
Malaria , Microbiota , Respiratory Distress Syndrome , Animals , Disease Models, Animal , Lung/pathology , Malaria/complications , Malaria/parasitology , Plasmodium berghei/physiology
5.
Int J Parasitol ; 52(11): 711-715, 2022 10.
Article in English | MEDLINE | ID: mdl-35367213

ABSTRACT

While the liver and blood stages of the Plasmodium life cycle are commonly regarded as two separate fields of malaria research, several studies have pointed towards the existence of a bidirectional cross-talk, where one stage of mammalian infection may impact the establishment and progression of the other. Despite the constraints in experimentally addressing concurrent liver and blood stage Plasmodium infections, animal models and clinical studies have unveiled a plethora of molecular interactions between the two. Here, we review the current knowledge on the reciprocal influence of hepatic and erythrocytic infection by malaria parasites, and discuss its impacts on immunity, pathology and vaccination against this deadly disease.


Subject(s)
Malaria , Parasites , Plasmodium , Animals , Malaria/parasitology , Liver/parasitology , Life Cycle Stages , Mammals
6.
iScience ; 23(12): 101781, 2020 Dec 18.
Article in English | MEDLINE | ID: mdl-33294789

ABSTRACT

Plasmodium parasites, causative agents of malaria, scavenge host nutrients to sustain their intracellular replication. Modulation of the host's nutritional status can potentially help control infection by limiting the parasite's access to nutrients, or by boosting the immune system. Here, we show that dietary supplementation of mice employing a combination of arginine (R) with two additional amino acids, lysine (K) and valine (V), termed RKV, significantly decreases Plasmodium liver infection. RKV supplementation results in the elimination of parasites at a late stage of their development in the liver. Our data employing genetic knockout mouse models and in vivo depletion of specific cell populations suggest that RKV supplementation boosts the host's overall innate immune response, and that parasite elimination is dependent on MyD88 signaling in immune cells. The immunostimulatory effect of RKV supplementation opens a potential role for dietary supplementation as an adjuvant for prophylaxis or immunization strategies against Plasmodium infection.

7.
Commun Biol ; 3(1): 688, 2020 11 19.
Article in English | MEDLINE | ID: mdl-33214643

ABSTRACT

The malaria parasite Plasmodium obligatorily infects and replicates inside hepatocytes surrounded by a parasitophorous vacuole membrane (PVM), which is decorated by the host-cell derived autophagy protein LC3. We have previously shown that the parasite-derived, PVM-resident protein UIS3 sequesters LC3 to avoid parasite elimination by autophagy from hepatocytes. Here we show that a small molecule capable of disrupting this interaction triggers parasite elimination in a host cell autophagy-dependent manner. Molecular docking analysis of more than 20 million compounds combined with a phenotypic screen identified one molecule, C4 (4-{[4-(4-{5-[3-(trifluoromethyl) phenyl]-1,2,4-oxadiazol-3-yl}benzyl)piperazino]carbonyl}benzonitrile), capable of impairing infection. Using biophysical assays, we established that this impairment is due to the ability of C4 to disrupt UIS3-LC3 interaction, thus inhibiting the parasite's ability to evade the host autophagy response. C4 impacts infection in autophagy-sufficient cells without harming the normal autophagy pathway of the host cell. This study, by revealing the disruption of a critical host-parasite interaction without affecting the host's normal function, uncovers an efficient anti-malarial strategy to prevent this deadly disease.


Subject(s)
Antimalarials/pharmacology , Membrane Proteins/metabolism , Plasmodium berghei/physiology , Plasmodium falciparum/physiology , Protozoan Proteins/metabolism , Animals , Antimalarials/therapeutic use , Autophagy , Cell Adhesion , Databases, Chemical , Humans , Malaria/drug therapy , Malaria/parasitology , Male , Membrane Proteins/chemistry , Mice , Mice, Inbred C57BL , Protein Binding , Protein Conformation , Protozoan Proteins/chemistry
8.
Trends Parasitol ; 36(1): 11-18, 2020 01.
Article in English | MEDLINE | ID: mdl-31787522

ABSTRACT

Plasmodium, the causative agent of malaria, is responsible for more than 200 million new infections and 400 000 deaths yearly. While in recent years the influence of the microbiota in homeostasis and a wide variety of disorders has taken center stage, its contribution during malaria infections has only now started to emerge. The few published studies suggest two distinct but complementary directions. Plasmodium infections can cause significant alterations in host (at least gut) microbiota, and host gut microbiota can influence the clinical outcome of malaria infections. In this opinion article, we highlight the most fundamental unanswered questions in the field that will, hopefully, point future research directions towards unveiling key mechanistic insights of the Plasmodium-host-microbiota axis.


Subject(s)
Host-Parasite Interactions , Malaria/microbiology , Microbiota/physiology , Animals , Gastrointestinal Microbiome/physiology , Humans , Plasmodium/physiology
9.
Nat Microbiol ; 2(12): 1600-1607, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28947801

ABSTRACT

The relevance of genetic factors in conferring protection to severe malaria has been demonstrated, as in the case of sickle cell trait and G6PD deficiency 1 . However, it remains unknown whether environmental components, such as dietary or metabolic variations, can contribute to the outcome of infection 2 . Here, we show that administration of a high-fat diet to mice for a period as short as 4 days impairs Plasmodium liver infection by over 90%. Plasmodium sporozoites can successfully invade and initiate replication but die inside hepatocytes, thereby are unable to cause severe disease. Transcriptional analyses combined with genetic and chemical approaches reveal that this impairment of infection is mediated by oxidative stress. We show that reactive oxygen species, probably spawned from fatty acid ß-oxidation, directly impact Plasmodium survival inside hepatocytes, and parasite load can be rescued by exogenous administration of the antioxidant N-acetylcysteine or the ß-oxidation inhibitor etomoxir. Together, these data reveal that acute and transient dietary alterations markedly impact the establishment of a Plasmodium infection and disease outcome.


Subject(s)
Diet, High-Fat/methods , Host-Parasite Interactions/genetics , Malaria/diet therapy , Acetylcysteine/metabolism , Animals , Antioxidants/metabolism , Disease Models, Animal , Female , Gene Expression Profiling , Glucose Tolerance Test , Glucosephosphate Dehydrogenase Deficiency/metabolism , Hep G2 Cells , Hepatocytes/metabolism , Hepatocytes/parasitology , Humans , Liver/metabolism , Liver/parasitology , Liver Diseases/metabolism , Liver Diseases/parasitology , Macrophages/parasitology , Macrophages/pathology , Malaria/blood , Malaria/pathology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress , Parasite Load , Plasmodium berghei , Reactive Oxygen Species , Sickle Cell Trait/metabolism , Sporozoites/metabolism
10.
Nat Med ; 20(1): 47-53, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24362933

ABSTRACT

Before they infect red blood cells and cause malaria, Plasmodium parasites undergo an obligate and clinically silent expansion phase in the liver that is supposedly undetected by the host. Here, we demonstrate the engagement of a type I interferon (IFN) response during Plasmodium replication in the liver. We identified Plasmodium RNA as a previously unrecognized pathogen-associated molecular pattern (PAMP) capable of activating a type I IFN response via the cytosolic pattern recognition receptor Mda5. This response, initiated by liver-resident cells through the adaptor molecule for cytosolic RNA sensors, Mavs, and the transcription factors Irf3 and Irf7, is propagated by hepatocytes in an interferon-α/ß receptor-dependent manner. This signaling pathway is critical for immune cell-mediated host resistance to liver-stage Plasmodium infection, which we find can be primed with other PAMPs, including hepatitis C virus RNA. Together, our results show that the liver has sensor mechanisms for Plasmodium that mediate a functional antiparasite response driven by type I IFN.


Subject(s)
Immunity, Innate/immunology , Interferon Type I/immunology , Liver/parasitology , Plasmodium/immunology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blotting, Western , DEAD-box RNA Helicases/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Profiling , Green Fluorescent Proteins , Immunohistochemistry , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/metabolism , Interferon-Induced Helicase, IFIH1 , Liver/immunology , Luciferases , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microarray Analysis , Oligonucleotides/genetics , Plasmodium/genetics , Real-Time Polymerase Chain Reaction , Statistics, Nonparametric
11.
Immunity ; 39(5): 874-84, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24184056

ABSTRACT

Severe sepsis remains a poorly understood systemic inflammatory condition with high mortality rates and limited therapeutic options in addition to organ support measures. Here we show that the clinically approved group of anthracyclines acts therapeutically at a low dose regimen to confer robust protection against severe sepsis in mice. This salutary effect is strictly dependent on the activation of DNA damage response and autophagy pathways in the lung, as demonstrated by deletion of the ataxia telangiectasia mutated (Atm) or the autophagy-related protein 7 (Atg7) specifically in this organ. The protective effect of anthracyclines occurs irrespectively of pathogen burden, conferring disease tolerance to severe sepsis. These findings demonstrate that DNA damage responses, including the ATM and Fanconi Anemia pathways, are important modulators of immune responses and might be exploited to confer protection to inflammation-driven conditions, including severe sepsis.


Subject(s)
Anthracyclines/pharmacology , Anti-Bacterial Agents/pharmacology , DNA Repair/drug effects , Lung/drug effects , Peritonitis/drug therapy , Sepsis/prevention & control , Adenoviridae Infections/immunology , Animals , Anthracyclines/therapeutic use , Anti-Bacterial Agents/therapeutic use , Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/physiology , Autophagy-Related Protein 7 , Cecum/injuries , DNA Damage , Epirubicin/administration & dosage , Epirubicin/pharmacology , Epirubicin/therapeutic use , Fanconi Anemia Complementation Group D2 Protein/physiology , Inflammation , Inflammation Mediators/analysis , Injections, Intraperitoneal , Lung/metabolism , Meropenem , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/physiology , Organ Specificity , Peritonitis/etiology , Peritonitis/genetics , Peritonitis/immunology , Peritonitis/physiopathology , Respiratory Tract Infections/immunology , Shock, Septic/prevention & control , Thienamycins/therapeutic use , Whole-Body Irradiation
12.
Eur J Immunol ; 42(7): 1843-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22585713

ABSTRACT

Effective CD8(+) T-cell responses against tumor or microbial antigens that are not directly expressed in antigen-presenting cells (APCs) depend on the cross-presentation of these antigens on MHC class I in APCs. To identify signaling molecules that regulate cross-presentation, we used lentiviral-based RNA interference to test the roles of hundreds of kinases and phosphatases in this process. Our study uncovered eight previously unknown genes, consisting of one positive and seven negative regulators of antigen cross-presentation. Depletion of Acvr1c, a type I receptor for TGF-ß family of signaling molecules, led to an increase in CD80 and CD86 co-stimulator surface expression and secreted IL-12 in mouse bone marrow-derived DCs, as well as antigen-specific T-cell proliferation.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/enzymology , Dendritic Cells/immunology , Phosphoric Monoester Hydrolases/immunology , Phosphotransferases/immunology , Activin Receptors, Type I/genetics , Activin Receptors, Type I/immunology , Animals , Antigen Presentation/genetics , Blotting, Western , Cross-Priming/genetics , Cross-Priming/immunology , Flow Cytometry , Gene Silencing/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphoric Monoester Hydrolases/genetics , Phosphotransferases/genetics , RNA/chemistry , RNA/genetics , RNA Interference/immunology
13.
PLoS One ; 6(5): e19829, 2011.
Article in English | MEDLINE | ID: mdl-21611201

ABSTRACT

The generation of diversity and plasticity of transcriptional programs are key components of effective vertebrate immune responses. The role of Alternative Splicing has been recognized, but it is underappreciated and poorly understood as a critical mechanism for the regulation and fine-tuning of physiological immune responses. Here we report the generation of loss-of-function phenotypes for a large collection of genes known or predicted to be involved in the splicing reaction and the identification of 19 novel regulators of IL-1ß secretion in response to E. coli challenge of THP-1 cells. Twelve of these genes are required for IL-1ß secretion, while seven are negative regulators of this process. Silencing of SFRS3 increased IL-1ß secretion due to elevation of IL-1ß and caspase-1 mRNA in addition to active caspase-1 levels. This study points to the relevance of splicing in the regulation of auto-inflammatory diseases.


Subject(s)
Interleukin-1beta/metabolism , RNA Splicing/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism , Caspase 1/metabolism , Cell Line , Enzyme Activation , Escherichia coli , Gene Expression Regulation , Gene Silencing , Genes, Reporter , Humans , Interleukin-1beta/genetics , Monocytes/metabolism , NF-kappa B/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Reproducibility of Results , Serine-Arginine Splicing Factors , Transcription, Genetic
14.
Cell ; 145(3): 398-409, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21529713

ABSTRACT

Sickle human hemoglobin (Hb) confers a survival advantage to individuals living in endemic areas of malaria, the disease caused by Plasmodium infection. As demonstrated hereby, mice expressing sickle Hb do not succumb to experimental cerebral malaria (ECM). This protective effect is exerted irrespectively of parasite load, revealing that sickle Hb confers host tolerance to Plasmodium infection. Sickle Hb induces the expression of heme oxygenase-1 (HO-1) in hematopoietic cells, via a mechanism involving the transcription factor NF-E2-related factor 2 (Nrf2). Carbon monoxide (CO), a byproduct of heme catabolism by HO-1, prevents further accumulation of circulating free heme after Plasmodium infection, suppressing the pathogenesis of ECM. Moreover, sickle Hb inhibits activation and/or expansion of pathogenic CD8(+) T cells recognizing antigens expressed by Plasmodium, an immunoregulatory effect that does not involve Nrf2 and/or HO-1. Our findings provide insight into molecular mechanisms via which sickle Hb confers host tolerance to severe forms of malaria.


Subject(s)
Hemoglobin, Sickle/immunology , Malaria/immunology , Plasmodium berghei , Animals , CD8-Positive T-Lymphocytes/immunology , Carbon Monoxide/metabolism , Chemokines/metabolism , Crosses, Genetic , Disease Models, Animal , Heme Oxygenase-1/metabolism , Host-Pathogen Interactions , Humans , Malaria/physiopathology , Malaria, Cerebral/immunology , Malaria, Cerebral/physiopathology , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism
15.
Sci Transl Med ; 2(51): 51ra71, 2010 Sep 29.
Article in English | MEDLINE | ID: mdl-20881280

ABSTRACT

Low-grade polymicrobial infection induced by cecal ligation and puncture is lethal in heme oxygenase-1-deficient mice (Hmox1(-/-)), but not in wild-type (Hmox1(+/+)) mice. Here we demonstrate that the protective effect of this heme-catabolizing enzyme relies on its ability to prevent tissue damage caused by the circulating free heme released from hemoglobin during infection. Heme administration after low-grade infection in mice promoted tissue damage and severe sepsis. Free heme contributed to the pathogenesis of severe sepsis irrespective of pathogen load, revealing that it compromised host tolerance to infection. Development of lethal forms of severe sepsis after high-grade infection was associated with reduced serum concentrations of the heme sequestering protein hemopexin (HPX), whereas HPX administration after high-grade infection prevented tissue damage and lethality. Finally, the lethal outcome of septic shock in patients was also associated with reduced HPX serum concentrations. We propose that targeting free heme by HPX might be used therapeutically to treat severe sepsis.


Subject(s)
Heme/metabolism , Sepsis/etiology , Sepsis/metabolism , Animals , Apoptosis , Bacterial Infections/enzymology , Bacterial Infections/immunology , HMGB1 Protein/metabolism , Heme Oxygenase-1/metabolism , Hemopexin/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Immune Tolerance/immunology , Mice , Models, Biological , Oxidation-Reduction , Sepsis/immunology , Sepsis/physiopathology
16.
Blood ; 114(19): 4064-76, 2009 Nov 05.
Article in English | MEDLINE | ID: mdl-19696201

ABSTRACT

Severe sepsis is one of the leading causes of death worldwide. High mortality rates in sepsis are frequently associated with neutropenia. Despite the central role of neutrophils in innate immunity, the mechanisms causing neutropenia during sepsis remain elusive. Here, we show that neutropenia is caused in part by apoptosis and is sustained by a block of hematopoietic stem cell (HSC) differentiation. Using a sepsis murine model, we found that the human opportunistic bacterial pathogen Pseudomonas aeruginosa caused neutrophil depletion and expansion of the HSC pool in the bone marrow. "Septic" HSCs were significantly impaired in competitive repopulation assays and defective in generating common myeloid progenitors and granulocyte-monocyte progenitors, resulting in lower rates of myeloid differentiation in vitro and in vivo. Delayed myeloid-neutrophil differentiation was further mapped using a lysozyme-green fluorescent protein (GFP) reporter mouse. Pseudomonas's lipopolysaccharide was necessary and sufficient to induce myelosuppresion and required intact TLR4 signaling. Our results establish a previously unrecognized link between HSC regulation and host response in severe sepsis and demonstrate a novel role for TLR4.


Subject(s)
Hematopoietic Stem Cells/pathology , Myeloid Cells/pathology , Sepsis/pathology , Animals , Apoptosis , Cell Differentiation/drug effects , Disease Models, Animal , Female , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Humans , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/pathology , Myeloid Cells/drug effects , Neutropenia/etiology , Neutropenia/immunology , Neutropenia/pathology , Pseudomonas Infections/complications , Pseudomonas Infections/immunology , Pseudomonas Infections/pathology , Sepsis/complications , Sepsis/immunology , Signal Transduction , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
17.
Proc Natl Acad Sci U S A ; 106(37): 15837-42, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19706490

ABSTRACT

Infection by Plasmodium, the causative agent of malaria, is associated with hemolysis and therefore with release of hemoglobin from RBC. Under inflammatory conditions, cell-free hemoglobin can be oxidized, releasing its heme prosthetic groups and producing deleterious free heme. Here we demonstrate that survival of a Plasmodium-infected host relies strictly on its ability to prevent the cytotoxic effects of free heme via the expression of the heme-catabolyzing enzyme heme oxygenase-1 (HO-1; encoded by the Hmox1 gene). When infected with Plasmodium chabaudi chabaudi (Pcc), wild-type (Hmox1(+/+)) BALB/c mice resolved infection and restored homeostasis thereafter (0% lethality). In contrast, HO-1 deficient (Hmox1(-/-)) BALB/c mice developed a lethal form of hepatic failure (100% lethality), similar to the one occurring in Pcc-infected DBA/2 mice (75% lethality). Expression of HO-1 suppresses the pro-oxidant effects of free heme, preventing it from sensitizing hepatocytes to undergo TNF-mediated programmed cell death by apoptosis. This cytoprotective effect, which inhibits the development of hepatic failure in Pcc-infected mice without interfering with pathogen burden, is mimicked by pharmacological antioxidants such as N-acetylcysteine (NAC). When administered therapeutically, i.e., after Pcc infection, NAC suppressed the development of hepatic failure in Pcc-infected DBA/2 mice (0% lethality), without interfering with pathogen burden. In conclusion, we describe a mechanism of host defense against Plasmodium infection, based on tissue cytoprotection against free heme and limiting disease severity irrespectively of parasite burden.


Subject(s)
Heme Oxygenase-1/metabolism , Malaria/enzymology , Malaria/prevention & control , Plasmodium chabaudi/pathogenicity , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Gene Expression , Heme/metabolism , Heme Oxygenase-1/deficiency , Heme Oxygenase-1/genetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Liver Failure/pathology , Liver Failure/prevention & control , Malaria/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Mice, Knockout , Mice, SCID , Oxidative Stress , Plasmodium chabaudi/physiology , Transplantation Chimera , Tumor Necrosis Factor-alpha/pharmacology
18.
J Biol Chem ; 284(43): 29582-95, 2009 Oct 23.
Article in English | MEDLINE | ID: mdl-19700768

ABSTRACT

Several pathologic conditions are associated with hemolysis, i.e. release of ferrous (Fe(II)) hemoglobin from red blood cells. Oxidation of cell-free hemoglobin produces (Fe(III)) methemoglobin. More extensive oxidation produces (Fe(III)/Fe(IV) O) ferryl hemoglobin. Both cell-free methemoglobin and ferryl hemoglobin are thought to contribute to the pathogenesis of hemolytic disorders. We show hereby that ferryl hemoglobin, but not hemoglobin or methemoglobin, acts as a potent proinflammatory agonist that induces vascular endothelial cells in vitro to rearrange the actin cytoskeleton, forming intercellular gaps and disrupting the integrity of the endothelial cell monolayer. Furthermore, ferryl hemoglobin induces the expression of proinflammatory genes in endothelial cells in vitro, e.g. E-selectin, Icam-1, and Vcam-1, through the activation of the nuclear factor kappaB family of transcription factors. This proinflammatory effect, which requires actin polymerization, involves the activation of the c-Jun N-terminal kinase and the p38 mitogen-activated protein kinase signal transduction pathways. When administered to naïve mice, ferryl hemoglobin induces the recruitment of polymorphonuclear cells, demonstrating that it acts as a proinflammatory agonist in vivo. In conclusion, oxidized hemoglobin, i.e. ferryl hemoglobin, acts as a proinflammatory agonist that targets vascular endothelial cells.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Regulation , Hemoglobins/metabolism , Inflammation Mediators/metabolism , MAP Kinase Signaling System , Oxyhemoglobins/metabolism , Animals , Cell Adhesion Molecules/biosynthesis , Cell Line , Hemoglobins/pharmacology , Hemolysis , Humans , Inflammation Mediators/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Methemoglobin/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , NF-kappa B/metabolism , Neutrophils/metabolism , Oxyhemoglobins/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
19.
Exp Hematol ; 36(5): 545-558, 2008 May.
Article in English | MEDLINE | ID: mdl-18439488

ABSTRACT

OBJECTIVE: Homeostasis of the hematopoietic compartment is challenged and maintained during conditions of stress by mechanisms that are poorly defined. To understand how the bone marrow (BM) microenvironment influences hematopoiesis, we explored the role of Notch signaling and BM endothelial cells in providing microenvironmental cues to hematopoietic cells in the presence of inflammatory stimuli. MATERIALS AND METHODS: The human BM endothelial cell line (BMEC) and primary human BM endothelial cells were analyzed for expression of Notch ligands and the ability to expand hematopoietic progenitors in an in vitro coculture system. In vivo experiments were carried out to identify modulation of Notch signaling in BM endothelial and hematopoietic cells in mice challenged with tumor necrosis factor-alpha (TNF-alpha) or lipopolysaccharide (LPS), or in Tie2-tmTNF-alpha transgenic mice characterized by constitutive TNF-alpha activation. RESULTS: BM endothelial cells were found to express Jagged ligands and to greatly support progenitor's colony-forming ability. This effect was markedly decreased by Notch antagonists and augmented by increasing levels of Jagged2. Physiologic upregulation of Jagged2 expression on BMEC was observed upon TNF-alpha activation. Injection of TNF-alpha or LPS upregulated three- to fourfold Jagged2 expression on murine BM endothelial cells in vivo and resulted in increased Notch activation on murine hematopoietic stem/progenitor cells. Similarly, constitutive activation of endothelial cells in Tie2-tmTNF-alpha mice was characterized by increased expression of Jagged2 and by augmented Notch activation on hematopoietic stem/progenitor cells. CONCLUSIONS: Our results provide the first evidence that BM endothelial cells promote expansion of hematopoietic progenitor cells by a Notch-dependent mechanism and that TNF-alpha and LPS can modulate the levels of Notch ligand expression and Notch activation in the BM microenvironment in vivo.


Subject(s)
Bone Marrow/immunology , Endothelial Cells/immunology , Inflammation/immunology , Receptors, Notch/metabolism , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/physiology , Animals , Bone Marrow/blood supply , Bone Marrow/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Hematopoietic Stem Cells/immunology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-2 Protein , Ligands , Lipopolysaccharides/pharmacology , Membrane Proteins/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred Strains , Mice, Transgenic , Receptors, Notch/drug effects , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , Up-Regulation/immunology
20.
Nat Med ; 13(6): 703-10, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17496899

ABSTRACT

Cerebral malaria claims more than 1 million lives per year. We report that heme oxygenase-1 (HO-1, encoded by Hmox1) prevents the development of experimental cerebral malaria (ECM). BALB/c mice infected with Plasmodium berghei ANKA upregulated HO-1 expression and activity and did not develop ECM. Deletion of Hmox1 and inhibition of HO activity increased ECM incidence to 83% and 78%, respectively. HO-1 upregulation was lower in infected C57BL/6 compared to BALB/c mice, and all infected C57BL/6 mice developed ECM (100% incidence). Pharmacological induction of HO-1 and exposure to the end-product of HO-1 activity, carbon monoxide (CO), reduced ECM incidence in C57BL/6 mice to 10% and 0%, respectively. Whereas neither HO-1 nor CO affected parasitemia, both prevented blood-brain barrier (BBB) disruption, brain microvasculature congestion and neuroinflammation, including CD8(+) T-cell brain sequestration. These effects were mediated by the binding of CO to hemoglobin, preventing hemoglobin oxidation and the generation of free heme, a molecule that triggers ECM pathogenesis.


Subject(s)
Carbon Monoxide/physiology , Heme Oxygenase-1/physiology , Heme/metabolism , Malaria, Cerebral/enzymology , Animals , Disease Models, Animal , Heme Oxygenase-1/deficiency , Heme Oxygenase-1/genetics , Malaria, Cerebral/drug therapy , Malaria, Cerebral/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Plasmodium berghei
SELECTION OF CITATIONS
SEARCH DETAIL
...