Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Más filtros










Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 112(42): 13075-80, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26438846

RESUMEN

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.


Asunto(s)
Antimaláricos/uso terapéutico , Diazooxonorleucina/uso terapéutico , Glutamina/metabolismo , Malaria Cerebral/tratamiento farmacológico , Malaria Falciparum/tratamiento farmacológico , Animales , Antimaláricos/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Diazooxonorleucina/farmacología , Malaria Cerebral/metabolismo , Malaria Falciparum/metabolismo , Ratones
2.
Infect Immun ; 83(11): 4229-36, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26283330

RESUMEN

Malaria elimination efforts would benefit from vaccines that block transmission of Plasmodium falciparum gametocytes from humans to mosquitoes. A clear understanding of gametocyte-specific antibody responses in exposed populations could help determine whether transmission-blocking vaccines (TBV) would be boosted by natural gametocyte exposure, and also inform the development of serologic tools to monitor gametocyte exposure in populations targeted for malaria elimination. To this end, plasma was collected from Malian children and adults before and after the 6-month malaria season and probed against a microarray containing 1,204 P. falciparum proteins. Using publicly available proteomic data, we classified 91 proteins as gametocyte specific and 69 as proteins not expressed by gametocytes. The overall breadth and magnitude of gametocyte-specific IgG responses increased during the malaria season, although they were consistently lower than IgG responses to nongametocyte antigens. Notably, IgG specific for the TBV candidates Pfs48/45 and Pfs230 increased during the malaria season. In addition, IgGs specific for the gametocyte proteins Pfmdv1, Pfs16, PF3D7_1346400, and PF3D7_1024800 were detected in nearly all subjects, suggesting that seroconversion to these proteins may be a sensitive indicator of gametocyte exposure, although further studies are needed to determine the specificity and kinetics of these potential serologic markers. These findings suggest that TBV-induced immunity would be boosted through natural gametocyte exposure, and that antibody responses to particular antigens may reliably indicate gametocyte exposure.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Células Germinativas/inmunología , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Adolescente , Adulto , Niño , Preescolar , Estudios de Cohortes , Femenino , Humanos , Lactante , Malaria Falciparum/parasitología , Masculino , Plasmodium falciparum/química , Plasmodium falciparum/clasificación , Plasmodium falciparum/genética , Proteómica , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Adulto Joven
3.
mBio ; 6(3): e00725, 2015 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-26037126

RESUMEN

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.


Asunto(s)
Malaria Cerebral/prevención & control , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Encéfalo/patología , Perfilación de la Expresión Génica , Malaria Cerebral/patología , Ratones , Análisis de Supervivencia
4.
J Immunol ; 194(3): 929-39, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25556245

RESUMEN

Immunity to Plasmodium falciparum malaria is naturally acquired in individuals living in malaria-endemic areas of Africa. Abs play a key role in mediating this immunity; however, the acquisition of the components of Ab immunity, long-lived plasma cells and memory B cells (MBCs), is remarkably inefficient, requiring years of malaria exposure. Although long-lived classical MBCs (CD19(+)/CD20(+)/CD21(+)/CD27(+)/CD10(-)) are gradually acquired in response to natural infection, exposure to P. falciparum also results in a large expansion of what we have termed atypical MBCs (CD19(+)/CD20(+)/CD21(-)/CD27(-)/CD10(-)). At present, the function of atypical MBCs in malaria is not known, nor are the factors that drive their differentiation. To gain insight into the relationship between classical and atypical IgG(+) MBCs, we compared the Ab H and L chain V gene repertoires of children living in a malaria-endemic region in Mali. We found that these repertoires were remarkably similar by a variety of criteria, including V gene usage, rate of somatic hypermutation, and CDR-H3 length and composition. The similarity in these repertoires suggests that classical MBCs and atypical MBCs differentiate in response to similar Ag-dependent selective pressures in malaria-exposed children and that atypical MBCs do not express a unique V gene repertoire.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Susceptibilidad a Enfermedades , Región Variable de Inmunoglobulina/genética , Memoria Inmunológica/genética , Malaria/genética , Malaria/inmunología , África Occidental , Secuencia de Aminoácidos , Antígenos de Protozoos/inmunología , Niño , Preescolar , Estudios de Cohortes , Regiones Determinantes de Complementariedad/genética , Femenino , Reordenamiento Génico de Linfocito B , Variación Genética , Humanos , Cadenas Pesadas de Inmunoglobulina/química , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/química , Cadenas Ligeras de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/química , Inmunofenotipificación , Malaria Falciparum/genética , Malaria Falciparum/inmunología , Masculino , Plasmodium falciparum/inmunología , Hipermutación Somática de Inmunoglobulina
5.
PLoS Pathog ; 10(9): e1004338, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25211214

RESUMEN

BACKGROUND: Invasion of mosquito salivary glands (SGs) by Plasmodium falciparum sporozoites is an essential step in the malaria life cycle. How infection modulates gene expression, and affects hematophagy remains unclear. PRINCIPAL FINDINGS: Using Affimetrix chip microarray, we found that at least 43 genes are differentially expressed in the glands of Plasmodium falciparum-infected Anopheles gambiae mosquitoes. Among the upregulated genes, one codes for Agaphelin, a 58-amino acid protein containing a single Kazal domain with a Leu in the P1 position. Agaphelin displays high homology to orthologs present in Aedes sp and Culex sp salivary glands, indicating an evolutionarily expanded family. Kinetics and surface plasmon resonance experiments determined that chemically synthesized Agaphelin behaves as a slow and tight inhibitor of neutrophil elastase (K(D) ∼ 10 nM), but does not affect other enzymes, nor promotes vasodilation, or exhibit antimicrobial activity. TAXIscan chamber assay revealed that Agaphelin inhibits neutrophil chemotaxis toward fMLP, affecting several parameter associated with cell migration. In addition, Agaphelin reduces paw edema formation and accumulation of tissue myeloperoxidase triggered by injection of carrageenan in mice. Agaphelin also blocks elastase/cathepsin-mediated platelet aggregation, abrogates elastase-mediated cleavage of tissue factor pathway inhibitor, and attenuates neutrophil-induced coagulation. Notably, Agaphelin inhibits neutrophil extracellular traps (NETs) formation and prevents FeCl3-induced arterial thrombosis, without impairing hemostasis. CONCLUSIONS: Blockade of neutrophil elastase emerges as a novel antihemostatic mechanism in hematophagy; it also supports the notion that neutrophils and the innate immune response are targets for antithrombotic therapy. In addition, Agaphelin is the first antihemostatic whose expression is induced by Plasmodium sp infection. These results suggest that an important interplay takes place in parasite-vector-host interactions.


Asunto(s)
Anopheles/parasitología , Hemostasis/fisiología , Interacciones Huésped-Parásitos , Proteínas de Insectos/metabolismo , Neutrófilos/inmunología , Plasmodium falciparum/patogenicidad , Proteínas y Péptidos Salivales/metabolismo , Trombosis/prevención & control , Secuencia de Aminoácidos , Animales , Anopheles/metabolismo , Dicroismo Circular , Edema/etiología , Edema/metabolismo , Edema/prevención & control , Femenino , Proteínas de Insectos/química , Proteínas de Insectos/genética , Insectos Vectores , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Glándulas Salivales/metabolismo , Glándulas Salivales/parasitología , Proteínas y Péptidos Salivales/química , Proteínas y Péptidos Salivales/genética , Homología de Secuencia de Aminoácido , Resonancia por Plasmón de Superficie
6.
PLoS One ; 9(2): e87140, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586264

RESUMEN

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.


Asunto(s)
Antioxidantes/farmacología , Óxidos N-Cíclicos/farmacología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Malaria Cerebral/tratamiento farmacológico , Tromboplastina/metabolismo , Animales , Antioxidantes/uso terapéutico , Células Cultivadas , Quimiocina CCL2/metabolismo , Óxidos N-Cíclicos/uso terapéutico , Ensayo de Inmunoadsorción Enzimática , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Malaria Cerebral/metabolismo , Ratones , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Marcadores de Spin
7.
Annu Rev Immunol ; 32: 157-87, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24655294

RESUMEN

Malaria is a mosquito-borne disease caused by parasites of the obligate intracellular Apicomplexa phylum the most deadly of which, Plasmodium falciparum, prevails in Africa. Malaria imposes a huge health burden on the world's most vulnerable populations, claiming the lives of nearly one million children and pregnant women each year. Although there is keen interest in eradicating malaria, we do not yet have the necessary tools to meet this challenge, including an effective malaria vaccine and adequate vector control strategies. Here we review what is known about the mechanisms at play in immune resistance to malaria in both the human and mosquito hosts at each step in the parasite's complex life cycle with a view toward developing the tools that will contribute to the prevention of disease and death and, ultimately, to the goal of malaria eradication. In so doing, we hope to inspire immunologists to participate in defeating this devastating disease.


Asunto(s)
Culicidae/inmunología , Interacciones Huésped-Patógeno/inmunología , Malaria/inmunología , Plasmodium/inmunología , Animales , Culicidae/parasitología , Humanos , Estadios del Ciclo de Vida , Malaria/parasitología , Malaria/prevención & control , Plasmodium/crecimiento & desarrollo , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/inmunología
8.
Mem Inst Oswaldo Cruz ; 108(6): 707-17, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24037192

RESUMEN

Schistosomiasis is a major neglected tropical disease caused by trematodes from the genus Schistosoma. Because schistosomes exhibit a complex life cycle and numerous mechanisms for regulating gene expression, it is believed that spliced leader (SL) trans-splicing could play an important role in the biology of these parasites. The purpose of this study was to investigate the function of trans-splicing in Schistosoma mansoni through analysis of genes that may be regulated by this mechanism and via silencing SL-containing transcripts through RNA interference. Here, we report our analysis of SL transcript-enriched cDNA libraries from different S. mansoni life stages. Our results show that the trans-splicing mechanism is apparently not associated with specific genes, subcellular localisations or life stages. In cross-species comparisons, even though the sets of genes that are subject to SL trans-splicing regulation appear to differ between organisms, several commonly shared orthologues were observed. Knockdown of trans-spliced transcripts in sporocysts resulted in a systemic reduction of the expression levels of all tested trans-spliced transcripts; however, the only phenotypic effect observed was diminished larval size. Further studies involving the findings from this work will provide new insights into the role of trans-splicing in the biology of S. mansoni and other organisms. All Expressed Sequence Tags generated in this study were submitted to dbEST as five different libraries. The accessions for each library and for the individual sequences are as follows: (i) adult worms of mixed sexes (LIBEST_027999: JZ139310 - JZ139779), (ii) female adult worms (LIBEST_028000: JZ139780 - JZ140379), (iii) male adult worms (LIBEST_028001: JZ140380 - JZ141002), (iv) eggs (LIBEST_028002: JZ141003 - JZ141497) and (v) schistosomula (LIBEST_028003: JZ141498 - JZ141974).


Asunto(s)
Técnicas de Silenciamiento del Gen , Precursores del ARN/aislamiento & purificación , ARN Lider Empalmado/genética , Schistosoma mansoni/genética , Trans-Empalme/fisiología , Animales , Etiquetas de Secuencia Expresada , Femenino , Regulación de la Expresión Génica/genética , Biblioteca de Genes , Larva , Estadios del Ciclo de Vida/genética , Masculino , Fenotipo , Precursores del ARN/genética , ARN Bicatenario , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Schistosoma mansoni/crecimiento & desarrollo , Trans-Empalme/genética
9.
Mem. Inst. Oswaldo Cruz ; 108(6): 707-717, set. 2013. tab, graf
Artículo en Inglés | LILACS | ID: lil-685497

RESUMEN

Schistosomiasis is a major neglected tropical disease caused by trematodes from the genus Schistosoma. Because schistosomes exhibit a complex life cycle and numerous mechanisms for regulating gene expression, it is believed that spliced leader (SL) trans-splicing could play an important role in the biology of these parasites. The purpose of this study was to investigate the function of trans-splicing in Schistosoma mansoni through analysis of genes that may be regulated by this mechanism and via silencing SL-containing transcripts through RNA interference. Here, we report our analysis of SL transcript-enriched cDNA libraries from different S. mansoni life stages. Our results show that the trans-splicing mechanism is apparently not associated with specific genes, subcellular localisations or life stages. In cross-species comparisons, even though the sets of genes that are subject to SL trans-splicing regulation appear to differ between organisms, several commonly shared orthologues were observed. Knockdown of trans-spliced transcripts in sporocysts resulted in a systemic reduction of the expression levels of all tested trans-spliced transcripts; however, the only phenotypic effect observed was diminished larval size. Further studies involving the findings from this work will provide new insights into the role of trans-splicing in the biology of S. mansoni and other organisms. All Expressed Sequence Tags generated in this study were submitted to dbEST as five different libraries. The accessions for each library and for the individual sequences are as follows: (i) adult worms of mixed sexes (LIBEST_027999: JZ139310 - JZ139779), (ii) female adult worms (LIBEST_028000: JZ139780 - JZ140379), (iii) male adult worms (LIBEST_028001: JZ140380 - JZ141002), (iv) eggs (LIBEST_028002: JZ141003 - JZ141497) and (v) schistosomula (LIBEST_028003: JZ141498 - JZ141974).


Asunto(s)
Animales , Femenino , Masculino , Técnicas de Silenciamiento del Gen , Precursores del ARN/aislamiento & purificación , ARN Lider Empalmado/genética , Schistosoma mansoni/genética , Trans-Empalme/fisiología , Etiquetas de Secuencia Expresada , Biblioteca de Genes , Regulación de la Expresión Génica/genética , Larva , Estadios del Ciclo de Vida/genética , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa , Precursores del ARN/genética , ARN Bicatenario , ARN Interferente Pequeño/metabolismo , Schistosoma mansoni/crecimiento & desarrollo , Trans-Empalme/genética
10.
PLoS One ; 8(5): e62820, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23675429

RESUMEN

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.


Asunto(s)
Predisposición Genética a la Enfermedad , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Malaria/genética , Malaria/inmunología , Placenta/parasitología , Complicaciones Parasitarias del Embarazo , Animales , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Índices de Eritrocitos , Femenino , Lupus Eritematoso Sistémico/complicaciones , Malaria/sangre , Malaria/complicaciones , Ratones , Parasitemia/genética , Parasitemia/inmunología , Placenta/patología , Embarazo , Reproducción/genética , Reproducción/inmunología , Índice de Severidad de la Enfermedad
11.
Proc Natl Acad Sci U S A ; 109(14): 5429-34, 2012 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-22431641

RESUMEN

The malaria parasite, Plasmodium falciparum, and the human immune system have coevolved to ensure that the parasite is not eliminated and reinfection is not resisted. This relationship is likely mediated through a myriad of host-parasite interactions, although surprisingly few such interactions have been identified. Here we show that the 33-kDa fragment of P. falciparum merozoite surface protein 1 (MSP1(33)), an abundant protein that is shed during red blood cell invasion, binds to the proinflammatory protein, S100P. MSP1(33) blocks S100P-induced NFκB activation in monocytes and chemotaxis in neutrophils. Remarkably, S100P binds to both dimorphic alleles of MSP1, estimated to have diverged >27 Mya, suggesting an ancient, conserved relationship between these parasite and host proteins that may serve to attenuate potentially damaging inflammatory responses.


Asunto(s)
Proteínas de Unión al Calcio/antagonistas & inhibidores , Proteína 1 de Superficie de Merozoito/fisiología , Proteínas de Neoplasias/antagonistas & inhibidores , Plasmodium falciparum/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de Unión al Calcio/química , Cromatografía en Gel , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Humanos , Microscopía Confocal , Datos de Secuencia Molecular , Proteínas de Neoplasias/química , Homología de Secuencia de Aminoácido , Resonancia por Plasmón de Superficie
12.
PLoS One ; 7(1): e29493, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22242171

RESUMEN

Malaria has had the largest impact of any infectious disease on shaping the human genome, exerting enormous selective pressure on genes that improve survival in severe malaria infections. Modern humans originated in Africa and lost skin melanization as they migrated to temperate regions of the globe. Although it is well documented that loss of melanization improved cutaneous Vitamin D synthesis, melanin plays an evolutionary ancient role in insect immunity to malaria and in some instances melanin has been implicated to play an immunoregulatory role in vertebrates. Thus, we tested the hypothesis that melanization may be protective in malaria infections using mouse models. Congenic C57BL/6 mice that differed only in the gene encoding tyrosinase, a key enzyme in the synthesis of melanin, showed no difference in the clinical course of infection by Plasmodium yoelii 17XL, that causes severe anemia, Plasmodium berghei ANKA, that causes severe cerebral malaria or Plasmodium chabaudi AS that causes uncomplicated chronic disease. Moreover, neither genetic deficiencies in vitamin D synthesis nor vitamin D supplementation had an effect on survival in cerebral malaria. Taken together, these results indicate that neither melanin nor vitamin D production improve survival in severe malaria.


Asunto(s)
Malaria/prevención & control , Melaninas/metabolismo , Modelos Biológicos , Animales , Enfermedad Crónica , Humanos , Malaria/complicaciones , Malaria/parasitología , Malaria Cerebral/complicaciones , Malaria Cerebral/parasitología , Malaria Cerebral/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Parasitemia/complicaciones , Parasitemia/tratamiento farmacológico , Plasmodium/fisiología , Receptores de Calcitriol/metabolismo , Vitamina D/uso terapéutico
13.
Arterioscler Thromb Vasc Biol ; 32(3): 786-98, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22116094

RESUMEN

OBJECTIVE: The coagulation-inflammation cycle has been implicated as a critical component in malaria pathogenesis. Defibrotide (DF), a mixture of DNA aptamers, displays anticoagulant, anti-inflammatory, and endothelial cell (EC)-protective activities and has been successfully used to treat comatose children with veno-occlusive disease. DF was investigated here as a drug to treat cerebral malaria. METHODS AND RESULTS: DF blocks tissue factor expression by ECs incubated with parasitized red blood cells and attenuates prothrombinase activity, platelet aggregation, and complement activation. In contrast, it does not affect nitric oxide bioavailability. We also demonstrated that Plasmodium falciparum glycosylphosphatidylinositol (Pf-GPI) induces tissue factor expression in ECs and cytokine production by dendritic cells. Notably, dendritic cells, known to modulate coagulation and inflammation systemically, were identified as a novel target for DF. Accordingly, DF inhibits Toll-like receptor ligand-dependent dendritic cells activation by a mechanism that is blocked by adenosine receptor antagonist (8-p-sulfophenyltheophylline) but not reproduced by synthetic poly-A, -C, -T, and -G. These results imply that aptameric sequences and adenosine receptor mediate dendritic cells responses to the drug. DF also prevents rosetting formation, red blood cells invasion by P. falciparum and abolishes oocysts development in Anopheles gambiae. In a murine model of cerebral malaria, DF affected parasitemia, decreased IFN-γ levels, and ameliorated clinical score (day 5) with a trend for increased survival. CONCLUSION: Therapeutic use of DF in malaria is proposed.


Asunto(s)
Antiinflamatorios/farmacología , Anticoagulantes/farmacología , Antimaláricos/farmacología , Coagulación Sanguínea/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Malaria Cerebral/tratamiento farmacológico , Plasmodium berghei/efectos de los fármacos , Plasmodium falciparum/efectos de los fármacos , Polidesoxirribonucleótidos/farmacología , Animales , Células Cultivadas , Activación de Complemento/efectos de los fármacos , Citocinas/sangre , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/parasitología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Células Endoteliales/parasitología , Femenino , Glicosilfosfatidilinositoles/metabolismo , Hemoglobinas/metabolismo , Humanos , Mediadores de Inflamación/sangre , Malaria Cerebral/sangre , Malaria Cerebral/inmunología , Malaria Cerebral/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Plasmodium berghei/patogenicidad , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidad , Agregación Plaquetaria/efectos de los fármacos , Receptores Purinérgicos P1/efectos de los fármacos , Receptores Purinérgicos P1/metabolismo , Índice de Severidad de la Enfermedad , Tromboplastina/metabolismo , Factores de Tiempo
14.
Nat Immunol ; 12(11): 1119-26, 2011 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-21964606

RESUMEN

Binding of antigen to the B cell antigen receptor (BCR) triggers both BCR signaling and endocytosis. How endocytosis regulates BCR signaling remains unknown. Here we report that BCR signaling was not extinguished by endocytosis of BCRs; instead, BCR signaling initiated at the plasma membrane continued as the BCR trafficked intracellularly with the sequential phosphorylation of kinases. Blocking the endocytosis of BCRs resulted in the recruitment of both proximal and downstream kinases to the plasma membrane, where mitogen-activated protein kinases (MAPKs) were hyperphosphorylated and the kinase Akt and its downstream target Foxo were hypophosphorylated, which led to the dysregulation of gene transcription controlled by these pathways. Thus, the cellular location of the BCR serves to compartmentalize kinase activation to regulate the outcome of signaling.


Asunto(s)
Linfocitos B/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Compartimento Celular/inmunología , Células Cultivadas , Endocitosis/inmunología , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Fosforilación/inmunología , Transporte de Proteínas/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Transducción de Señal/inmunología , Activación Transcripcional/inmunología
15.
J Infect Dis ; 204(11): 1750-61, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21998476

RESUMEN

BACKGROUND: Heterozygous states of hemoglobin (Hb) A and HbS (HbAS, sickle-cell trait) or HbC (HbAC) protect against Plasmodium falciparum malaria by unclear mechanisms. Several studies suggest that HbAS and HbAC accelerate the acquisition of immunity to malaria, possibly by enhancing P. falciparum-specific antibody responses. METHODS: We used a protein microarray representing 491 P. falciparum proteins expressed during exoerythrocytic and erythrocytic stages of the life cycle to test the hypothesis that HbAS and HbAC enhance the P. falciparum-specific IgG response compared with normal HbAA. Plasma samples were collected from Malian children aged 2-10 years before and after a 6-month malaria season and were probed against the microarray. Immunoglobulin G (IgG) profiles of children with HbAA (n = 106), HbAS (n = 15), and HbAC (n = 20) were compared. RESULTS: Although the magnitude and breadth of P. falciparum-specific IgG responses increased with age and from before to after the malaria season in each antigen category, Hb type did not independently predict significant differences in P. falciparum-specific IgG profiles. CONCLUSIONS: These data do not support the hypothesis that HbAS and HbAC protect against malaria by enhancing P. falciparum-specific antibody responses. It remains possible that HbAS and HbAC protect against malaria by enhancing antibody responses to antigens not studied here or through other immune mechanisms.


Asunto(s)
Anticuerpos Antiprotozoarios/sangre , Antígenos de Protozoos/inmunología , Hemoglobina C/inmunología , Hemoglobina Falciforme/inmunología , Inmunoglobulina G/sangre , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Factores de Edad , Animales , Niño , Preescolar , Femenino , Hemoglobina A/inmunología , Hemoglobina C/genética , Hemoglobina Falciforme/genética , Heterocigoto , Humanos , Estadios del Ciclo de Vida/inmunología , Malaria Falciparum/parasitología , Masculino , Malí , Plasmodium falciparum/crecimiento & desarrollo , Análisis por Matrices de Proteínas
16.
Proc Natl Acad Sci U S A ; 108(3): 1122-7, 2011 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-21187399

RESUMEN

Plasmodium falciparum has exerted tremendous selective pressure on genes that improve survival in severe malarial infections. Systemic lupus erythematosus (SLE) is an autoimmune disease that is six to eight times more prevalent in women of African descent than in women of European descent. Here we provide evidence that a genetic susceptibility to SLE protects against cerebral malaria. Mice that are prone to SLE because of a deficiency in FcγRIIB or overexpression of Toll-like receptor 7 are protected from death caused by cerebral malaria. Protection appears to be by immune mechanisms that allow SLE-prone mice better to control their overall inflammatory responses to parasite infections. These findings suggest that the high prevalence of SLE in women of African descent living outside of Africa may result from the inheritance of genes that are beneficial in the immune control of cerebral malaria but that, in the absence of malaria, contribute to autoimmune disease.


Asunto(s)
Población Negra/genética , Predisposición Genética a la Enfermedad/genética , Lupus Eritematoso Sistémico/genética , Malaria Cerebral/genética , Plasmodium berghei/inmunología , Receptores de IgG/deficiencia , Receptor Toll-Like 7/metabolismo , Animales , Encéfalo/inmunología , Encéfalo/patología , Citocinas/sangre , Cartilla de ADN/genética , Ensayo de Inmunoadsorción Enzimática , Eritrocitos/parasitología , Femenino , Citometría de Flujo , Humanos , Lupus Eritematoso Sistémico/etnología , Malaria Cerebral/inmunología , Malaria Cerebral/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos , Receptores de IgG/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/fisiología , Análisis de Supervivencia
17.
Proc Natl Acad Sci U S A ; 107(15): 6958-63, 2010 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-20351286

RESUMEN

Abs are central to malaria immunity, which is only acquired after years of exposure to Plasmodium falciparum (Pf). Despite the enormous worldwide burden of malaria, the targets of protective Abs and the basis of their inefficient acquisition are unknown. Addressing these knowledge gaps could accelerate malaria vaccine development. To this end, we developed a protein microarray containing approximately 23% of the Pf 5,400-protein proteome and used this array to probe plasma from 220 individuals between the ages of 2-10 years and 18-25 years in Mali before and after the 6-month malaria season. Episodes of malaria were detected by passive surveillance over the 8-month study period. Ab reactivity to Pf proteins rose dramatically in children during the malaria season; however, most of this response appeared to be short-lived based on cross-sectional analysis before the malaria season, which revealed only modest incremental increases in Ab reactivity with age. Ab reactivities to 49 Pf proteins measured before the malaria season were significantly higher in 8-10-year-old children who were infected with Pf during the malaria season but did not experience malaria (n = 12) vs. those who experienced malaria (n = 29). This analysis also provided insight into patterns of Ab reactivity against Pf proteins based on the life cycle stage at which proteins are expressed, subcellular location, and other proteomic features. This approach, if validated in larger studies and in other epidemiological settings, could prove to be a useful strategy for better understanding fundamental properties of the human immune response to Pf and for identifying previously undescribed vaccine targets.


Asunto(s)
Malaria Falciparum/inmunología , Plasmodium falciparum/metabolismo , Análisis por Matrices de Proteínas/métodos , Adolescente , Adulto , Animales , Antígenos de Protozoos/inmunología , Niño , Preescolar , Estudios de Cohortes , Humanos , Sistema Inmunológico , Vacunas contra la Malaria/química , Malí , Proteómica/métodos
18.
Nat Genet ; 42(3): 268-71, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20101240

RESUMEN

Antimalarial drugs impose strong selective pressure on Plasmodium falciparum parasites and leave signatures of selection in the parasite genome; screening for genes under selection may suggest potential drug or immune targets. Genome-wide association studies (GWAS) of parasite traits have been hampered by the lack of high-throughput genotyping methods, inadequate knowledge of parasite population history and time-consuming adaptations of parasites to in vitro culture. Here we report the first Plasmodium GWAS, which included 189 culture-adapted P. falciparum parasites genotyped using a custom-built Affymetrix molecular inversion probe 3K malaria panel array with a coverage of approximately 1 SNP per 7 kb. Population structure, variation in recombination rate and loci under recent positive selection were detected. Parasite half-maximum inhibitory concentrations for seven antimalarial drugs were obtained and used in GWAS to identify genes associated with drug responses. This study provides valuable tools and insight into the P. falciparum genome.


Asunto(s)
Antimaláricos/uso terapéutico , Resistencia a Medicamentos/genética , Plasmodium falciparum/genética , Recombinación Genética , Selección Genética , Antimaláricos/farmacología , Mapeo Cromosómico , Análisis por Conglomerados , Hibridación Genómica Comparativa/métodos , ADN Protozoario/análisis , Sitios Genéticos , Estudio de Asociación del Genoma Completo , Geografía , Concentración 50 Inhibidora , Análisis de Secuencia por Matrices de Oligonucleótidos , Recombinación Genética/efectos de los fármacos , Selección Genética/efectos de los fármacos
19.
BMC Genomics ; 8: 181, 2007 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-17578584

RESUMEN

BACKGROUND: The parasitic trematode Schistosoma mansoni is one of the major causative agents of Schistosomiasis, a disease that affects approximately 200 million people, mostly in developing countries. Since much of the pathology is associated with eggs laid by the female worm, understanding the mechanisms involved in oogenesis and sexual maturation is an important step towards the discovery of new targets for effective drug therapy. It is known that the adult female worm only develops fully in the presence of a male worm and that the rates of oviposition and maturation of eggs are significantly increased by mating. In order to study gene transcripts associated with sexual maturation and oviposition, we compared the gene expression profiles of sexually mature and immature parasites using DNA microarrays. RESULTS: For each experiment, three amplified RNA microarray hybridizations and their dye swaps were analyzed. Our results show that 265 transcripts are differentially expressed in adult females and 53 in adult males when mature and immature worms are compared. Of the genes differentially expressed, 55% are expressed at higher levels in paired females while the remaining 45% are more expressed in unpaired ones and 56.6% are expressed at higher levels in paired male worms while the remaining 43.4% are more expressed in immature parasites. Real-time RT-PCR analysis validated the microarray results. Several new maturation associated transcripts were identified. Genes that were up-regulated in single-sex females were mostly related to energy generation (i.e. carbohydrate and protein metabolism, generation of precursor metabolites and energy, cellular catabolism, and organelle organization and biogenesis) while genes that were down-regulated related to RNA metabolism, reactive oxygen species metabolism, electron transport, organelle organization and biogenesis and protein biosynthesis. CONCLUSION: Our results confirm previous observations related to gene expression induced by sexual maturation in female schistosome worms. They also increase the list of S. mansoni maturation associated transcripts considerably, therefore opening new and exciting avenues for the study of the conjugal biology and development of new drugs against schistosomes.


Asunto(s)
Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Schistosoma mansoni/genética , Conducta Sexual Animal , Animales , Clonación Molecular , ADN Complementario , Femenino , Genes de Helminto , Masculino , Modelos Genéticos , Hibridación de Ácido Nucleico , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Toxicology ; 192(2-3): 95-117, 2003 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-14580780

RESUMEN

Cadmium is a heavy metal, which is widely used in industry, affecting human health through occupational and environmental exposure. In mammals, it exerts multiple toxic effects and has been classified as a human carcinogen by the International Agency for Research on Cancer. Cadmium affects cell proliferation, differentiation, apoptosis and other cellular activities. Cd2+ does not catalyze Fenton-type reactions because it does not accept or donate electrons under physiological conditions, and it is only weakly genotoxic. Hence, indirect mechanisms are implicated in the carcinogenicity of cadmium. In this review multiple mechanisms are discussed, such as modulation of gene expression and signal transduction, interference with enzymes of the cellular antioxidant system and generation of reactive oxygen species (ROS), inhibition of DNA repair and DNA methylation, role in apoptosis and disruption of E-cadherin-mediated cell-cell adhesion. Cadmium affects both gene transcription and translation. The major mechanisms of gene induction by cadmium known so far are modulation of cellular signal transduction pathways by enhancement of protein phosphorylation and activation of transcription and translation factors. Cadmium interferes with antioxidant defense mechanisms and stimulates the production of reactive oxygen species, which may act as signaling molecules in the induction of gene expression and apoptosis. The inhibition of DNA repair processes by cadmium represents a mechanism by which cadmium enhances the genotoxicity of other agents and may contribute to the tumor initiation by this metal. The disruption of E-cadherin-mediated cell-cell adhesion by cadmium probably further stimulates the development of tumors. It becomes clear that there exist multiple mechanisms which contribute to the carcinogenicity of cadmium, although the relative weights of these contributions are difficult to estimate.


Asunto(s)
Cadmio/toxicidad , Carcinógenos Ambientales/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Neoplasias/inducido químicamente , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Humanos , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...