Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Immunity ; 56(3): 592-605.e8, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36804959

RESUMEN

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.


Asunto(s)
Eritropoyesis , Malaria Cerebral , Animales , Ratones , Eritrocitos , Interleucina-17 , Hígado/parasitología , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T gamma-delta , Malaria
2.
Nature ; 611(7936): 563-569, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36352220

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica , Hepatocitos , Hígado , Malaria , Parásitos , Plasmodium berghei , Análisis de la Célula Individual , Animales , Hepatocitos/citología , Hepatocitos/inmunología , Hepatocitos/metabolismo , Hepatocitos/parasitología , Hígado/anatomía & histología , Hígado/citología , Hígado/inmunología , Hígado/parasitología , Malaria/genética , Malaria/inmunología , Malaria/parasitología , Parásitos/genética , Parásitos/inmunología , Parásitos/metabolismo , Plasmodium berghei/genética , Plasmodium berghei/inmunología , Plasmodium berghei/metabolismo , Imagen Individual de Molécula , Análisis de Secuencia de ARN , Hierro/metabolismo , Ácidos Grasos/metabolismo , Transcripción Genética , Genes Protozoarios/genética , Interacciones Huésped-Parásitos/genética , Interacciones Huésped-Parásitos/inmunología
3.
Cell ; 145(3): 398-409, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21529713

RESUMEN

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.


Asunto(s)
Hemoglobina Falciforme/inmunología , Malaria/inmunología , Plasmodium berghei , Animales , Linfocitos T CD8-positivos/inmunología , Monóxido de Carbono/metabolismo , Quimiocinas/metabolismo , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Hemo-Oxigenasa 1/metabolismo , Interacciones Huésped-Patógeno , Humanos , Malaria/fisiopatología , Malaria Cerebral/inmunología , Malaria Cerebral/fisiopatología , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/metabolismo
4.
Immunity ; 39(5): 874-84, 2013 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-24184056

RESUMEN

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.


Asunto(s)
Antraciclinas/farmacología , Antibacterianos/farmacología , Reparación del ADN/efectos de los fármacos , Pulmón/efectos de los fármacos , Peritonitis/tratamiento farmacológico , Sepsis/prevención & control , Infecciones por Adenoviridae/inmunología , Animales , Antraciclinas/uso terapéutico , Antibacterianos/uso terapéutico , Proteínas de la Ataxia Telangiectasia Mutada/deficiencia , Proteínas de la Ataxia Telangiectasia Mutada/fisiología , Proteína 7 Relacionada con la Autofagia , Ciego/lesiones , Daño del ADN , Epirrubicina/administración & dosificación , Epirrubicina/farmacología , Epirrubicina/uso terapéutico , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/fisiología , Inflamación , Mediadores de Inflamación/análisis , Inyecciones Intraperitoneales , Pulmón/metabolismo , Meropenem , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/fisiología , Especificidad de Órganos , Peritonitis/etiología , Peritonitis/genética , Peritonitis/inmunología , Peritonitis/fisiopatología , Infecciones del Sistema Respiratorio/inmunología , Choque Séptico/prevención & control , Tienamicinas/uso terapéutico , Irradiación Corporal Total
5.
Nat Med ; 13(6): 703-10, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17496899

RESUMEN

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.


Asunto(s)
Monóxido de Carbono/fisiología , Hemo-Oxigenasa 1/fisiología , Hemo/metabolismo , Malaria Cerebral/enzimología , Animales , Modelos Animales de Enfermedad , Hemo-Oxigenasa 1/deficiencia , Hemo-Oxigenasa 1/genética , Malaria Cerebral/tratamiento farmacológico , Malaria Cerebral/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Plasmodium berghei
6.
Eur J Immunol ; 42(7): 1843-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22585713

RESUMEN

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.


Asunto(s)
Presentación de Antígeno/inmunología , Células Dendríticas/enzimología , Células Dendríticas/inmunología , Monoéster Fosfórico Hidrolasas/inmunología , Fosfotransferasas/inmunología , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/inmunología , Animales , Presentación de Antígeno/genética , Western Blotting , Reactividad Cruzada/genética , Reactividad Cruzada/inmunología , Citometría de Flujo , Silenciador del Gen/inmunología , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Monoéster Fosfórico Hidrolasas/genética , Fosfotransferasas/genética , ARN/química , ARN/genética , Interferencia de ARN/inmunología
7.
Proc Natl Acad Sci U S A ; 106(37): 15837-42, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19706490

RESUMEN

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.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Malaria/enzimología , Malaria/prevención & control , Plasmodium chabaudi/patogenicidad , Acetilcisteína/farmacología , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Expresión Génica , Hemo/metabolismo , Hemo-Oxigenasa 1/deficiencia , Hemo-Oxigenasa 1/genética , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Fallo Hepático/patología , Fallo Hepático/prevención & control , Malaria/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Ratones Noqueados , Ratones SCID , Estrés Oxidativo , Plasmodium chabaudi/fisiología , Quimera por Trasplante , Factor de Necrosis Tumoral alfa/farmacología
8.
Int J Parasitol ; 52(11): 711-715, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35367213

RESUMEN

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.


Asunto(s)
Malaria , Parásitos , Plasmodium , Animales , Malaria/parasitología , Hígado/parasitología , Estadios del Ciclo de Vida , Mamíferos
9.
Nat Commun ; 13(1): 3747, 2022 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-35768411

RESUMEN

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.


Asunto(s)
Malaria , Microbiota , Síndrome de Dificultad Respiratoria , Animales , Modelos Animales de Enfermedad , Pulmón/patología , Malaria/complicaciones , Malaria/parasitología , Plasmodium berghei/fisiología
10.
Elife ; 112022 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-36476511

RESUMEN

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.


Asunto(s)
Antraciclinas , FN-kappa B , Animales , Ratones , Antraciclinas/farmacología , Antibióticos Antineoplásicos/farmacología , Doxorrubicina/farmacología , Daño del ADN , ADN
11.
Blood ; 114(19): 4064-76, 2009 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-19696201

RESUMEN

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.


Asunto(s)
Células Madre Hematopoyéticas/patología , Células Mieloides/patología , Sepsis/patología , Animales , Apoptosis , Diferenciación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre Multipotentes/efectos de los fármacos , Células Madre Multipotentes/patología , Células Mieloides/efectos de los fármacos , Neutropenia/etiología , Neutropenia/inmunología , Neutropenia/patología , Infecciones por Pseudomonas/complicaciones , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/patología , Sepsis/complicaciones , Sepsis/inmunología , Transducción de Señal , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
12.
J Biol Chem ; 284(43): 29582-95, 2009 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-19700768

RESUMEN

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.


Asunto(s)
Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Hemoglobinas/metabolismo , Mediadores de Inflamación/metabolismo , Sistema de Señalización de MAP Quinasas , Oxihemoglobinas/metabolismo , Animales , Moléculas de Adhesión Celular/biosíntesis , Línea Celular , Hemoglobinas/farmacología , Hemólisis , Humanos , Mediadores de Inflamación/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Metahemoglobina/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , FN-kappa B/metabolismo , Neutrófilos/metabolismo , Oxihemoglobinas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
J Clin Invest ; 117(2): 438-47, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17256058

RESUMEN

Heme oxygenase-1 (HO-1, encoded by HMOX1) dampens inflammatory reactions via the catabolism of heme into CO, Fe, and biliverdin. We report that expression of HO-1 dictates the pathologic outcome of experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis (MS). Induction of EAE in Hmox1(-/- )C57BL/6 mice led to enhanced CNS demyelination, paralysis, and mortality, as compared with Hmox1(+/+) mice. Induction of HO-1 by cobalt protoporphyrin IX (CoPPIX) administration after EAE onset reversed paralysis in C57BL/6 and SJL/J mice and disease relapse in SJL/J mice. These effects were not observed using zinc protoporphyrin IX, which does not induce HO-1. CoPPIX protection was abrogated in Hmox1(-/-) C57BL/6 mice, indicating that CoPPIX acts via HO-1 to suppress EAE progression. The protective effect of HO-1 was associated with inhibition of MHC class II expression by APCs and inhibition of Th and CD8 T cell accumulation, proliferation, and effector function within the CNS. Exogenous CO mimicked these effects, suggesting that CO contributes to the protective action of HO-1. In conclusion, HO-1 or exposure to its end product CO counters autoimmune neuroinflammation and thus might be used therapeutically to treat MS.


Asunto(s)
Monóxido de Carbono/farmacología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Hemo-Oxigenasa 1/metabolismo , Animales , Células Presentadoras de Antígenos/inmunología , Autoinmunidad , Monóxido de Carbono/metabolismo , Encefalomielitis Autoinmune Experimental/prevención & control , Inducción Enzimática , Hemo-Oxigenasa 1/deficiencia , Hemo-Oxigenasa 1/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subgrupos de Linfocitos T/inmunología
14.
Trends Parasitol ; 36(1): 11-18, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31787522

RESUMEN

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.


Asunto(s)
Interacciones Huésped-Parásitos , Malaria/microbiología , Microbiota/fisiología , Animales , Microbioma Gastrointestinal/fisiología , Humanos , Plasmodium/fisiología
15.
Commun Biol ; 3(1): 688, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33214643

RESUMEN

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.


Asunto(s)
Antimaláricos/farmacología , Proteínas de la Membrana/metabolismo , Plasmodium berghei/fisiología , Plasmodium falciparum/fisiología , Proteínas Protozoarias/metabolismo , Animales , Antimaláricos/uso terapéutico , Autofagia , Adhesión Celular , Bases de Datos de Compuestos Químicos , Humanos , Malaria/tratamiento farmacológico , Malaria/parasitología , Masculino , Proteínas de la Membrana/química , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Conformación Proteica , Proteínas Protozoarias/química
16.
iScience ; 23(12): 101781, 2020 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-33294789

RESUMEN

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.

17.
Exp Hematol ; 36(5): 545-558, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18439488

RESUMEN

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.


Asunto(s)
Médula Ósea/inmunología , Células Endoteliales/inmunología , Inflamación/inmunología , Receptores Notch/metabolismo , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Médula Ósea/irrigación sanguínea , Médula Ósea/efectos de los fármacos , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Madre Hematopoyéticas/inmunología , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-2 , Ligandos , Lipopolisacáridos/farmacología , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos , Ratones Transgénicos , Receptores Notch/efectos de los fármacos , Receptores Notch/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
18.
Nat Microbiol ; 2(12): 1600-1607, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28947801

RESUMEN

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.


Asunto(s)
Dieta Alta en Grasa/métodos , Interacciones Huésped-Parásitos/genética , Malaria/dietoterapia , Acetilcisteína/metabolismo , Animales , Antioxidantes/metabolismo , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Deficiencia de Glucosafosfato Deshidrogenasa/metabolismo , Células Hep G2 , Hepatocitos/metabolismo , Hepatocitos/parasitología , Humanos , Hígado/metabolismo , Hígado/parasitología , Hepatopatías/metabolismo , Hepatopatías/parasitología , Macrófagos/parasitología , Macrófagos/patología , Malaria/sangre , Malaria/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Carga de Parásitos , Plasmodium berghei , Especies Reactivas de Oxígeno , Rasgo Drepanocítico/metabolismo , Esporozoítos/metabolismo
19.
FASEB J ; 19(3): 458-60, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15640283

RESUMEN

Heme oxygenase-1 (HO-1), which degrades heme into three products (carbon monoxide, free iron, and biliverdin), plays a protective role in many models of disease via its anti-inflammatory, anti-apoptotic, and anti-proliferative actions. Overexpression of HO-1 has been shown to suppress immune responses and prolong the survival of allografts; however, the underlying mechanism is not clear. We demonstrate two "new" properties of HO-1 that mediate activation induced cell death (AICD) of allo-antigen-responsive murine CD4+ T cells, resulting in immunomodulation. First, it functions in vivo and in vitro to "boost" the proliferative response of CD4+ T cells to allo-antigens in the early phase of allo-antigen-driven immune responses. This "boosting" effect is accompanied with a significant increase of activation markers and IL-2 production. Second, it exerts a pro-apoptotic effect in those activated T cells after the initial burst of proliferation. We further show that the AICD effect is mediated through the Fas/CD95-FasL signal transduction pathway. Correlating with the above-mentioned findings is the observed prolongation of mouse heart graft survival when HO-1 is expressed in vivo in both donor and recipient. In conclusion, induction of HO-1 expression accelerates clonal deletion of peripheral alloreactive CD4+ T cells by promoting AICD, which is presumably a key mechanism for its immunomodulatory effects such as in prolonging the survival of transplanted organs.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Muerte Celular , Hemo Oxigenasa (Desciclizante)/fisiología , Isoantígenos/inmunología , Activación de Linfocitos/fisiología , Animales , Apoptosis , Linfocitos T CD4-Positivos/enzimología , Inducción Enzimática/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Hemo Oxigenasa (Desciclizante)/biosíntesis , Hemo Oxigenasa (Desciclizante)/genética , Hemo-Oxigenasa 1 , Histocitoquímica , Inmunohistoquímica , Interleucina-2/análisis , Interleucina-2/metabolismo , Prueba de Cultivo Mixto de Linfocitos , Masculino , Proteínas de la Membrana , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Mutantes , Ratones Transgénicos , Ovalbúmina/genética , ARN Mensajero/análisis , Receptor fas/análisis
20.
Nat Med ; 20(1): 47-53, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24362933

RESUMEN

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.


Asunto(s)
Inmunidad Innata/inmunología , Interferón Tipo I/inmunología , Hígado/parasitología , Plasmodium/inmunología , Transducción de Señal/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Western Blotting , ARN Helicasas DEAD-box/inmunología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes , Inmunohistoquímica , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Helicasa Inducida por Interferón IFIH1 , Hígado/inmunología , Luciferasas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Análisis por Micromatrices , Oligonucleótidos/genética , Plasmodium/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Estadísticas no Paramétricas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA