Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(23): e2403796121, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38809710

RESUMEN

Olfactory receptors (Olfr) are G protein-coupled receptors that are normally expressed on olfactory sensory neurons to detect volatile chemicals or odorants. Interestingly, many Olfrs are also expressed in diverse tissues and function in cell-cell recognition, migration, and proliferation as well as immune responses and disease processes. Here, we showed that many Olfr genes were expressed in the mouse spleen, linked to Plasmodium yoelii genetic loci significantly, and/or had genome-wide patterns of LOD scores (GPLSs) similar to those of host Toll-like receptor genes. Expression of specific Olfr genes such as Olfr1386 in HEK293T cells significantly increased luciferase signals driven by IFN-ß and NF-κB promoters, with elevated levels of phosphorylated TBK1, IRF3, P38, and JNK. Mice without Olfr1386 were generated using the CRISPR/Cas9 method, and the Olfr1386-/- mice showed significantly lower IFN-α/ß levels and longer survival than wild-type (WT) littermates after infection with P. yoelii YM parasites. Inhibition of G protein signaling and P38 activity could affect cyclic AMP-responsive element promoter-driven luciferase signals and IFN-ß mRNA levels in HEK293T cells expressing the Olfr1386 gene, respectively. Screening of malaria parasite metabolites identified nicotinamide adenine dinucleotide (NAD) as a potential ligand for Olfr1386, and NAD could stimulate IFN-ß responses and phosphorylation of TBK1 and STAT1/2 in RAW264.7 cells. Additionally, parasite RNA (pRNA) could significantly increase Olfr1386 mRNA levels. This study links multiple Olfrs to host immune response pathways, identifies a candidate ligand for Olfr1386, and demonstrates the important roles of Olfr1386 in regulating type I interferon (IFN-I) responses during malaria parasite infections.


Asunto(s)
Interferón Tipo I , Malaria , Plasmodium yoelii , Receptores Odorantes , Animales , Ratones , Malaria/inmunología , Malaria/parasitología , Malaria/metabolismo , Humanos , Células HEK293 , Receptores Odorantes/genética , Receptores Odorantes/metabolismo , Interferón Tipo I/metabolismo , Interferón Tipo I/inmunología , Ratones Noqueados , Transducción de Señal , Ratones Endogámicos C57BL
2.
Proc Natl Acad Sci U S A ; 120(40): e2311557120, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-37748059

RESUMEN

Plasmodium parasites cause malaria with disease outcomes ranging from mild illness to deadly complications such as severe malarial anemia (SMA), pulmonary edema, acute renal failure, and cerebral malaria. In young children, SMA often requires blood transfusion and is a major cause of hospitalization. Malaria parasite infection leads to the destruction of infected and noninfected erythrocytes as well as dyserythropoiesis; however, the mechanism of dyserythropoiesis accompanied by splenomegaly is not completely understood. Using Plasmodium yoelii yoelii 17XNL as a model, we show that both a defect in erythroblastic island (EBI) macrophages in supporting red blood cell (RBC) maturation and the destruction of reticulocytes/RBCs by the parasites contribute to SMA and splenomegaly. After malaria parasite infection, the destruction of both infected and noninfected RBCs stimulates extramedullary erythropoiesis in mice. The continuous decline of RBCs stimulates active erythropoiesis and drives the expansion of EBIs in the spleen, contributing to splenomegaly. Phagocytosis of malaria parasites by macrophages in the bone marrow and spleen may alter their functional properties and abilities to support erythropoiesis, including reduced expression of the adherence molecule CD169 and inability to support erythroblast differentiation, particularly RBC maturation in vitro and in vivo. Therefore, macrophage dysfunction is a key mechanism contributing to SMA. Mitigating and/or alleviating the inhibition of RBC maturation may provide a treatment strategy for SMA.


Asunto(s)
Anemia , Malaria Cerebral , Plasmodium yoelii , Niño , Humanos , Animales , Ratones , Preescolar , Eritropoyesis , Esplenomegalia , Eritrocitos , Macrófagos
3.
Immunity ; 45(5): 1093-1107, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27793594

RESUMEN

Type I interferon (IFN) is critical for controlling pathogen infection; however, its regulatory mechanisms in plasmacytoid cells (pDCs) still remain unclear. Here, we have shown that nucleic acid sensors cGAS-, STING-, MDA5-, MAVS-, or transcription factor IRF3-deficient mice produced high amounts of type I IFN-α and IFN-ß (IFN-α/ß) in the serum and were resistant to lethal plasmodium yoelii YM infection. Robust IFN-α/ß production was abolished when gene encoding nucleic acid sensor TLR7, signaling adaptor MyD88, or transcription factor IRF7 was ablated or pDCs were depleted. Further, we identified SOCS1 as a key negative regulator to inhibit MyD88-dependent type I IFN signaling in pDCs. Finally, we have demonstrated that pDCs, cDCs, and macrophages were required for generating IFN-α/ß-induced subsequent protective immunity. Thus, our findings have identified a critical regulatory mechanism of type I IFN signaling in pDCs and stage-specific function of immune cells in generating potent immunity against lethal YM infection.


Asunto(s)
Inmunidad Adaptativa/inmunología , Células Dendríticas/inmunología , Interferón Tipo I/inmunología , Malaria/inmunología , Transducción de Señal/inmunología , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Técnicas de Silenciamiento del Gen , Ratones , Ratones Noqueados , Plasmodium yoelii , Reacción en Cadena de la Polimerasa
4.
Immunity ; 42(6): 1130-42, 2015 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-26070484

RESUMEN

Tissue-infiltrating Ly6C(hi) monocytes play diverse roles in immunity, ranging from pathogen killing to immune regulation. How and where this diversity of function is imposed remains poorly understood. Here we show that during acute gastrointestinal infection, priming of monocytes for regulatory function preceded systemic inflammation and was initiated prior to bone marrow egress. Notably, natural killer (NK) cell-derived IFN-γ promoted a regulatory program in monocyte progenitors during development. Early bone marrow NK cell activation was controlled by systemic interleukin-12 (IL-12) produced by Batf3-dependent dendritic cells (DCs) in the mucosal-associated lymphoid tissue (MALT). This work challenges the paradigm that monocyte function is dominantly imposed by local signals after tissue recruitment, and instead proposes a sequential model of differentiation in which monocytes are pre-emptively educated during development in the bone marrow to promote their tissue-specific function.


Asunto(s)
Células de la Médula Ósea/inmunología , Células Dendríticas/inmunología , Mucosa Intestinal/inmunología , Células Asesinas Naturales/inmunología , Leucocitos Mononucleares/inmunología , Toxoplasma/inmunología , Toxoplasmosis/inmunología , Animales , Antígenos Ly/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Células de la Médula Ósea/parasitología , Diferenciación Celular , Células Cultivadas , Interferón gamma/metabolismo , Interleucina-12/genética , Interleucina-12/metabolismo , Mucosa Intestinal/parasitología , Células Asesinas Naturales/parasitología , Leucocitos Mononucleares/parasitología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Inmunológicos , Especificidad de Órganos/inmunología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
5.
Antimicrob Agents Chemother ; 67(2): e0082122, 2023 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-36625569

RESUMEN

Protein ubiquitination is an important posttranslational regulation mechanism that mediates Plasmodium development and modifies parasite responses to antimalarial drugs. Although mutations in several parasite ubiquitination enzymes have been linked to increased drug tolerance, the molecular mechanisms by which ubiquitination pathways mediate these parasite responses remain largely unknown. Here, we investigate the roles of a Plasmodium falciparum ring finger ubiquitin ligase (PfRFUL) in parasite development and in responses to antimalarial drugs. We engineered a transgenic parasite having the Pfrful gene tagged with an HA-2A-NeoR-glmS sequence to knockdown (KD) Pfrful expression using glucosamine (GlcN). A Western blot analysis of the proteins from GlcN-treated pSLI-HA-NeoR-glmS-tagged (PfRFULg) parasites, relative to their wild-type (Dd2) controls, showed changes in the ubiquitination of numerous proteins. PfRFUL KD rendered the parasites more sensitive to multiple antimalarial drugs, including mefloquine, piperaquine, amodiaquine, and dihydroartemisinin. PfRFUL KD also decreased the protein level of the P. falciparum multiple drug resistance 1 protein (PfMDR1) and altered the ratio of two bands of the P. falciparum chloroquine resistance transporter (PfCRT), suggesting contributions to the changed drug responses by the altered ubiquitination of these two molecules. The inhibition of proteasomal protein degradation by epoxomicin increased the PfRFUL level, suggesting the degradation of PfRFUL by the proteasome pathways, whereas the inhibition of E3 ubiquitin ligase activities by JNJ26854165 reduced the PfRFUL level. This study reveals the potential mechanisms of PfRFUL in modifying the expression of drug transporters and their roles in parasite drug responses. PfRFUL could be a potential target for antimalarial drug development.


Asunto(s)
Antimaláricos , Plasmodium falciparum , Proteínas Protozoarias , Ubiquitina-Proteína Ligasas , Humanos , Antimaláricos/farmacología , Cloroquina/farmacología , Resistencia a Medicamentos/genética , Malaria Falciparum/tratamiento farmacológico , Proteínas de Transporte de Membrana/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
6.
Proc Natl Acad Sci U S A ; 117(32): 19465-19474, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32709745

RESUMEN

Infection by malaria parasites triggers dynamic immune responses leading to diverse symptoms and pathologies; however, the molecular mechanisms responsible for these reactions are largely unknown. We performed Trans-species Expression Quantitative Trait Locus analysis to identify a large number of host genes that respond to malaria parasite infections. Here we functionally characterize one of the host genes called receptor transporter protein 4 (RTP4) in responses to malaria parasite and virus infections. RTP4 is induced by type I IFN (IFN-I) and binds to the TANK-binding kinase (TBK1) complex where it negatively regulates TBK1 signaling by interfering with expression and phosphorylation of both TBK1 and IFN regulatory factor 3. Rtp4-/- mice were generated and infected with malaria parasite Plasmodiun berghei ANKA. Significantly higher levels of IFN-I response in microglia, lower parasitemia, fewer neurologic symptoms, and better survival rates were observed in Rtp4-/- than in wild-type mice. Similarly, RTP4 deficiency significantly reduced West Nile virus titers in the brain, but not in the heart and the spleen, of infected mice, suggesting a specific role for RTP4 in brain infection and pathology. This study reveals functions of RTP4 in IFN-I response and a potential target for therapy in diseases with neuropathology.


Asunto(s)
Encéfalo/patología , Interferón Tipo I/metabolismo , Malaria Cerebral/patología , Chaperonas Moleculares/metabolismo , Animales , Encéfalo/parasitología , Encéfalo/virología , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Factor 3 Regulador del Interferón , Malaria Cerebral/metabolismo , Malaria Cerebral/parasitología , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Chaperonas Moleculares/genética , Fosforilación , Plasmodium berghei/fisiología , Plasmodium yoelii/fisiología , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Fiebre del Nilo Occidental/metabolismo , Fiebre del Nilo Occidental/patología , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/fisiología
7.
Proc Natl Acad Sci U S A ; 117(28): 16567-16578, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32606244

RESUMEN

Malaria infection induces complex and diverse immune responses. To elucidate the mechanisms underlying host-parasite interaction, we performed a genetic screen during early (24 h) Plasmodium yoelii infection in mice and identified a large number of interacting host and parasite genes/loci after transspecies expression quantitative trait locus (Ts-eQTL) analysis. We next investigated a host E3 ubiquitin ligase gene (March1) that was clustered with interferon (IFN)-stimulated genes (ISGs) based on the similarity of the genome-wide pattern of logarithm of the odds (LOD) scores (GPLS). March1 inhibits MAVS/STING/TRIF-induced type I IFN (IFN-I) signaling in vitro and in vivo. However, in malaria-infected hosts, deficiency of March1 reduces IFN-I production by activating inhibitors such as SOCS1, USP18, and TRIM24 and by altering immune cell populations. March1 deficiency increases CD86+DC (dendritic cell) populations and levels of IFN-γ and interleukin 10 (IL-10) at day 4 post infection, leading to improved host survival. T cell depletion reduces IFN-γ level and reverse the protective effects of March1 deficiency, which can also be achieved by antibody neutralization of IFN-γ. This study reveals functions of MARCH1 (membrane-associated ring-CH-type finger 1) in innate immune responses and provides potential avenues for activating antimalaria immunity and enhancing vaccine efficacy.


Asunto(s)
Malaria/inmunología , Plasmodium yoelii/fisiología , Linfocitos T/inmunología , Ubiquitina-Proteína Ligasas/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Parásitos , Humanos , Inmunidad Innata , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Malaria/enzimología , Malaria/genética , Malaria/parasitología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasmodium yoelii/inmunología , Ubiquitina-Proteína Ligasas/genética
8.
Malar J ; 21(1): 333, 2022 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-36380373

RESUMEN

BACKGROUND: To understand how Plasmodium falciparum malaria is controlled, it is essential to elucidate the transcriptomic responses of the human host in naturally-exposed populations. Various individual studies of the human transcriptomic responses to naturally transmitted P. falciparum infections have been reported with varying results. Multicohort gene expression analysis by aggregating data from diverse populations into a single analysis will increase the reproducibility and reliability of the results. METHODS: In this study, discovery cohorts GSE1124-GPL96, GSE34404, GSE117613, and validation cohort GSE35858 were obtained from the Gene Expression Omnibus. A meta-analysis using data from the multicohort studies was performed to identify the differentially expressed genes (DEGs) between malaria-infected and noninfected individuals using the MetaIntegrator R package. Subsequently, the protein-protein interaction (PPI) networks of the DEGs were constructed using Cytoscape software. Significant modules were selected, and the hub genes were identified using the CytoHubba and MCODE plug-ins. Multicohort WGCNA was conducted to find a correlation between modules and malaria infection. Furthermore, the immune cell profile of the peripheral blood in different groups was identified using ssGSEA. RESULTS: These analyses reveal that neutrophil activation, neutrophil-mediated immunity, and neutrophil degranulation are involved in the human response to natural malaria infection. However, neutrophil cell enrichment and activation were not significantly different between mild malaria and severe malaria groups. Malaria infection also downregulates host genes in ribosome synthesis and protein translation and upregulates host cell division-related genes. Furthermore, immune cell profiling analysis shows that activated dendritic cells and type 2 T helper cells are upregulated, while activated B cells, immature B cells, and monocytes are downregulated in the malaria-infected patients relative to the noninfected individuals. Significantly higher enrichment of activated dendritic cell-related genes and significantly lower enrichment of monocyte-related genes are also observed in the peripheral blood of the severe malaria group than in the mild malaria group. CONCLUSION: These results reveal important molecular signatures of host responses to malaria infections, providing some bases for developing malaria control strategies and protective vaccines.


Asunto(s)
Malaria Falciparum , Malaria , Humanos , Plasmodium falciparum/genética , Reproducibilidad de los Resultados , Perfilación de la Expresión Génica , Transcriptoma
9.
BMC Genomics ; 22(1): 303, 2021 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-33902452

RESUMEN

BACKGROUND: Rodent malaria parasites are important models for studying host-malaria parasite interactions such as host immune response, mechanisms of parasite evasion of host killing, and vaccine development. One of the rodent malaria parasites is Plasmodium yoelii, and multiple P. yoelii strains or subspecies that cause different disease phenotypes have been widely employed in various studies. The genomes and transcriptomes of several P. yoelii strains have been analyzed and annotated, including the lethal strains of P. y. yoelii YM (or 17XL) and non-lethal strains of P. y. yoelii 17XNL/17X. Genomic DNA sequences and cDNA reads from another subspecies P. y. nigeriensis N67 have been reported for studies of genetic polymorphisms and parasite response to drugs, but its genome has not been assembled and annotated. RESULTS: We performed genome sequencing of the N67 parasite using the PacBio long-read sequencing technology, de novo assembled its genome and transcriptome, and predicted 5383 genes with high overall annotation quality. Comparison of the annotated genome of the N67 parasite with those of YM and 17X parasites revealed a set of genes with N67-specific orthology, expansion of gene families, particularly the homologs of the Plasmodium chabaudi erythrocyte membrane antigen, large numbers of SNPs and indels, and proteins predicted to interact with host immune responses based on their functional domains. CONCLUSIONS: The genomes of N67 and 17X parasites are highly diverse, having approximately one polymorphic site per 50 base pairs of DNA. The annotated N67 genome and transcriptome provide searchable databases for fast retrieval of genes and proteins, which will greatly facilitate our efforts in studying the parasite biology and gene function and in developing effective control measures against malaria.


Asunto(s)
Malaria , Parásitos , Plasmodium yoelii , Animales , Plasmodium yoelii/genética , Roedores , Transcriptoma
10.
Proc Natl Acad Sci U S A ; 115(49): 12513-12518, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30455312

RESUMEN

Concerns about malaria parasite resistance to treatment with artemisinin drugs (ARTs) have grown with findings of prolonged parasite clearance t1/2s (>5 h) and their association with mutations in Plasmodium falciparum Kelch-propeller protein K13. Here, we describe a P. falciparum laboratory cross of K13 C580Y mutant with C580 wild-type parasites to investigate ART response phenotypes in vitro and in vivo. After genotyping >400 isolated progeny, we evaluated 20 recombinants in vitro: IC50 measurements of dihydroartemisinin were at similar low nanomolar levels for C580Y- and C580-type progeny (mean ratio, 1.00; 95% CI, 0.62-1.61), whereas, in a ring-stage survival assay, the C580Y-type progeny had 19.6-fold (95% CI, 9.76-39.2) higher average counts. In splenectomized Aotus monkeys treated with three daily doses of i.v. artesunate, t1/2 calculations by three different methods yielded mean differences of 0.01 h (95% CI, -3.66 to 3.67), 0.80 h (95% CI, -0.92 to 2.53), and 2.07 h (95% CI, 0.77-3.36) between C580Y and C580 infections. Incidences of recrudescence were 57% in C580Y (4 of 7) versus 70% in C580 (7 of 10) infections (-13% difference; 95% CI, -58% to 35%). Allelic substitution of C580 in a C580Y-containing progeny clone (76H10) yielded a transformant (76H10C580Rev) that, in an infected monkey, recrudesced regularly 13 times over 500 d. Frequent recrudescences of ART-treated P. falciparum infections occur with or without K13 mutations and emphasize the need for improved partner drugs to effectively eliminate the parasites that persist through the ART component of combination therapy.


Asunto(s)
Antimaláricos/farmacología , Artemisininas/farmacología , Malaria Falciparum/parasitología , Plasmodium falciparum/efectos de los fármacos , Animales , Aotidae , Cruzamientos Genéticos , Resistencia a Medicamentos , Regulación de la Expresión Génica , Mutación , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
11.
Clin Microbiol Rev ; 32(4)2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31366610

RESUMEN

Protozoan Plasmodium parasites are the causative agents of malaria, a deadly disease that continues to afflict hundreds of millions of people every year. Infections with malaria parasites can be asymptomatic, with mild or severe symptoms, or fatal, depending on many factors such as parasite virulence and host immune status. Malaria can be treated with various drugs, with artemisinin-based combination therapies (ACTs) being the first-line choice. Recent advances in genetics and genomics of malaria parasites have contributed greatly to our understanding of parasite population dynamics, transmission, drug responses, and pathogenesis. However, knowledge gaps in parasite biology and host-parasite interactions still remain. Parasites resistant to multiple antimalarial drugs have emerged, while advanced clinical trials have shown partial efficacy for one available vaccine. Here we discuss genetic and genomic studies of Plasmodium biology, host-parasite interactions, population structures, mosquito infectivity, antigenic variation, and targets for treatment and immunization. Knowledge from these studies will advance our understanding of malaria pathogenesis, epidemiology, and evolution and will support work to discover and develop new medicines and vaccines.


Asunto(s)
Antimaláricos/farmacología , Resistencia a Medicamentos/genética , Evolución Molecular , Genoma de Protozoos/genética , Malaria/epidemiología , Malaria/parasitología , Plasmodium/efectos de los fármacos , Plasmodium/genética , Humanos , Plasmodium/clasificación , Plasmodium/patogenicidad
12.
Genome Res ; 26(9): 1288-99, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27531718

RESUMEN

The malaria parasite Plasmodium falciparum has a great capacity for evolutionary adaptation to evade host immunity and develop drug resistance. Current understanding of parasite evolution is impeded by the fact that a large fraction of the genome is either highly repetitive or highly variable and thus difficult to analyze using short-read sequencing technologies. Here, we describe a resource of deep sequencing data on parents and progeny from genetic crosses, which has enabled us to perform the first genome-wide, integrated analysis of SNP, indel and complex polymorphisms, using Mendelian error rates as an indicator of genotypic accuracy. These data reveal that indels are exceptionally abundant, being more common than SNPs and thus the dominant mode of polymorphism within the core genome. We use the high density of SNP and indel markers to analyze patterns of meiotic recombination, confirming a high rate of crossover events and providing the first estimates for the rate of non-crossover events and the length of conversion tracts. We observe several instances of meiotic recombination within copy number variants associated with drug resistance, demonstrating a mechanism whereby fitness costs associated with resistance mutations could be compensated and greater phenotypic plasticity could be acquired.


Asunto(s)
Resistencia a Medicamentos/genética , Variación Genética , Malaria Falciparum/genética , Plasmodium falciparum/genética , Mapeo Cromosómico , Variaciones en el Número de Copia de ADN/genética , Genoma de Protozoos/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mutación INDEL , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/parasitología , Meiosis/genética , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/patogenicidad , Polimorfismo de Nucleótido Simple , Recombinación Genética/genética
13.
Clin Infect Dis ; 67(11): 1670-1676, 2018 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-29846536

RESUMEN

Background: Mass drug administration (MDA), with or without low-dose primaquine (PMQLD), is being considered for malaria elimination programs. The potential of PMQLD to block malaria transmission by mosquitoes must be balanced against liabilities of its use. Methods: Artemisinin-piperaquine (AP), with or without PMQLD, was administered in 3 monthly rounds across Anjouan Island, Union of Comoros. Plasmodium falciparum malaria rates, mortality, parasitemias, adverse events, and PfK13 Kelch-propeller gene polymorphisms were evaluated. Results: Coverage of 85 to 93% of the Anjouan population was achieved with AP plus PMQLD (AP+PMQLD) in 2 districts (population 97164) and with AP alone in 5 districts (224471). Between the months of April-September in both 2012 and 2013, average monthly malaria hospital rates per 100000 people fell from 310.8 to 2.06 in the AP+PMQLD population (ratio 2.06/310.8 = 0.66%; 95% CI: 0.02%, 3.62%; P = .00007) and from 412.1 to 2.60 in the AP population (ratio 0.63%; 95% CI: 0.11%, 1.93%; P < .00001). Effectiveness of AP+PMQLD was 0.9908 (95% CI: 0.9053, 0.9991), while effectiveness of AP alone was 0.9913 (95% CI: 0.9657, 0.9978). Both regimens were well tolerated, without severe adverse events. Analysis of 52 malaria samples after MDA showed no evidence for selection of PfK13 Kelch-propeller mutations. Conclusions: Steep reductions of malaria cases were achieved by 3 monthly rounds of either AP+PMQLD or AP alone, suggesting potential for highly successful MDA without PMQLD in epidemiological settings such as those on Anjouan. A major challenge is to sustain and expand the public health benefits of malaria reductions by MDA.


Asunto(s)
Antimaláricos/uso terapéutico , Artemisininas/uso terapéutico , Malaria Falciparum/prevención & control , Primaquina/uso terapéutico , Quinolinas/uso terapéutico , Adolescente , Adulto , Niño , Preescolar , Comoras/epidemiología , ADN Protozoario/genética , Quimioterapia Combinada , Enfermedades Endémicas/prevención & control , Femenino , Humanos , Lactante , Malaria Falciparum/epidemiología , Malaria Falciparum/mortalidad , Masculino , Administración Masiva de Medicamentos , Parasitemia/tratamiento farmacológico , Parasitemia/epidemiología , Plasmodium falciparum , Polimorfismo Genético , Resultado del Tratamiento , Adulto Joven
14.
PLoS Pathog ; 12(10): e1005930, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27716849

RESUMEN

Both type I interferon (IFN-I) and CD40 play a significant role in various infectious diseases, including malaria and autoimmune disorders. CD40 is mostly known to function in adaptive immunity, but previous observations of elevated CD40 levels early after malaria infection of mice led us to investigate its roles in innate IFN-I responses and disease control. Using a Plasmodium yoelii nigeriensis N67 and C57BL/6 mouse model, we showed that infected CD40-/- mice had reduced STING and serum IFN-ß levels day-2 post infection, higher day-4 parasitemia, and earlier deaths. CD40 could greatly enhance STING-stimulated luciferase signals driven by the IFN-ß promoter in vitro, which was mediated by increased STING protein levels. The ability of CD40 to influence STING expression was confirmed in CD40-/- mice after malaria infection. Substitutions at CD40 TRAF binding domains significantly decreased the IFN-ß signals and STING protein level, which was likely mediated by changes in STING ubiquitination and degradation. Increased levels of CD40, STING, and ISRE driven luciferase signal in RAW Lucia were observed after phagocytosis of N67-infected red blood cells (iRBCs), stimulation with parasite DNA/RNA, or with selected TLR ligands [LPS, poly(I:C), and Pam3CSK4]. The results suggest stimulation of CD40 expression by parasite materials through TLR signaling pathways, which was further confirmed in bone marrow derived dendritic cells/macrophages (BMDCs/BMDMs) and splenic DCs from CD40-/-, TLR3-/- TLR4-/-, TRIF-/-, and MyD88-/- mice after iRBC stimulation or parasite infection. Our data connect several signaling pathways consisting of phagocytosis of iRBCs, recognition of parasite DNA/RNA (possibly GPI) by TLRs, elevated levels of CD40 and STING proteins, increased IFN-I production, and longer host survival time. This study reveals previously unrecognized CD40 function in innate IFN-I responses and protective pathways in infections with malaria strains that induce a strong IFN-I response, which may provide important information for better understanding and management of malaria.


Asunto(s)
Antígenos CD40/inmunología , Interacciones Huésped-Parásitos/inmunología , Interferón Tipo I/inmunología , Malaria/inmunología , Proteínas de la Membrana/inmunología , Animales , Western Blotting , Antígenos CD40/biosíntesis , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Células HEK293 , Humanos , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasmodium yoelii/inmunología
15.
Pharmacol Res ; 133: 77-100, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29727708

RESUMEN

The World Health Organization estimated that more than 1.5 billion people are infected with soil-transmitted helminths globally, and foodborne trematodiases in humans cause ∼2 million life-years lost to disability and death worldwide every year. Investment in prevention, treatment, and awareness of helminth infections and discovery of new, safe, effective, and affordable anti-helminth drugs are urgently needed. Artemisinin (ART) and its derivatives have been widely used to treat malaria and other protozoan infections; they also possess activities against helminths. So far, many papers on ART and its derivatives against schistosomal infections have been reported and reviewed. This review attempts to summarize recent advances in the uses of ART and its derivatives to treat infections of helminth parasites other than Schistosoma spp. in both humans and animals, including nematodes (Toxocara canis, Trichinella spiralis, Haemonchus contortus, Meloidogyne spp., Globodera rostochiensis, and Xiphinema index), cestodes (Echinococcus spp. and Taenia crassiceps), trematodes (Echinostoma spp., Fasciola spp., Clonorchis sinensis, Opisthorchis viverrini, Paragonimus westermani, Heterophyes heterophyes, and Paramphistomum microbothrium), and monogenea parasites (Dactylogyrus and Gyrodactylus). We concluded that ART and its derivatives are potentially effective drugs for treating various helminthic diseases of public health significance.


Asunto(s)
Antihelmínticos/uso terapéutico , Artemisininas/uso terapéutico , Helmintiasis/tratamiento farmacológico , Animales , Antihelmínticos/química , Antihelmínticos/farmacología , Artemisininas/química , Artemisininas/farmacología , Helmintos/efectos de los fármacos , Humanos
16.
Malar J ; 17(1): 83, 2018 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-29458365

RESUMEN

BACKGROUND: Malaria is still one of the serious public health problems in Grande Comore Island, although the number of annual cases has been greatly reduced in recent years. A better understanding of malaria parasite population diversity and transmission dynamics is critical for assessing the effectiveness of malaria control measures. The objective of this study is to investigate temporal changes in genetic diversity of Plasmodium falciparum populations and multiplicity of infection (MOI) in Grande Comore 10 years after introduction of ACT. METHODS: A total of 232 P. falciparum clinical isolates were collected from the Grande Comore Island during two sampling periods (118 for 2006‒2007 group, and 114 for 2013‒2016 group). Parasite isolates were characterized for genetic diversity and complexity of infection by genotyping polymorphic regions in merozoite surface protein gene 1 (msp-1), msp-2, and msp-3 using nested PCR and DNA sequencing. RESULTS: Three msp-1 alleles (K1, MAD20, and RO33), two msp-2 alleles (FC27 and 3D7), and two msp-3 alleles (K1 and 3D7) were detected in parasites of both sampling periods. The RO33 allele of msp-1 (84.8%), 3D7 allele of msp-2 (90.8%), and K1 allele of msp-3 (66.7%) were the predominant allelic types in isolates from 2006-2007 group. In contrast, the RO33 allele of msp-1 (63.4%), FC27 allele of msp-2 (91.1%), and 3D7 allele of msp-3 (53.5%) were the most prevalent among isolates from the 2013-2016 group. Compared with the 2006‒2007 group, polyclonal infection rates of msp-1 (from 76.7 to 29.1%, P < 0.01) and msp-2 (from 62.4 to 28.3%, P < 0.01) allelic types were significantly decreased in those from 2013‒2016 group. Similarly, the MOIs for both msp-1 and msp-2 were higher in P. falciparum isolates in the 2006-2007 group than those in 2013-2016 group (MOI = 3.11 vs 1.63 for msp-1; MOI = 2.75 vs 1.35 for msp-2). DNA sequencing analyses also revealed reduced numbers of distinct sequence variants in the three genes from 2006‒2007 to 2013‒2016: msp-1, from 32 to 23 (about 28% decline); msp-2 from 29 to 21 (about 28% decline), and msp-3 from 11 to 3 (about 72% decline). CONCLUSIONS: The present data showed dramatic reduction in genetic diversity and MOI among Grande Comore P. falciparum populations over the course of the study, suggesting a trend of decreasing malaria transmission intensity and genetic diversity in Grande Comore Island. These data provide valuable information for surveillance of P. falciparum infection and for assessing the appropriateness of the current malarial control strategies in the endemic area.


Asunto(s)
Antígenos de Protozoos/genética , Antimaláricos/uso terapéutico , Artemisininas/uso terapéutico , Quimioterapia Combinada/estadística & datos numéricos , Variación Genética , Proteína 1 de Superficie de Merozoito/genética , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Comoras , Humanos
17.
Nature ; 487(7407): 375-9, 2012 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-22722859

RESUMEN

Malaria elimination strategies require surveillance of the parasite population for genetic changes that demand a public health response, such as new forms of drug resistance. Here we describe methods for the large-scale analysis of genetic variation in Plasmodium falciparum by deep sequencing of parasite DNA obtained from the blood of patients with malaria, either directly or after short-term culture. Analysis of 86,158 exonic single nucleotide polymorphisms that passed genotyping quality control in 227 samples from Africa, Asia and Oceania provides genome-wide estimates of allele frequency distribution, population structure and linkage disequilibrium. By comparing the genetic diversity of individual infections with that of the local parasite population, we derive a metric of within-host diversity that is related to the level of inbreeding in the population. An open-access web application has been established for the exploration of regional differences in allele frequency and of highly differentiated loci in the P. falciparum genome.


Asunto(s)
Biodiversidad , Secuenciación de Nucleótidos de Alto Rendimiento , Malaria Falciparum/parasitología , Plasmodium falciparum/genética , Alelos , Genoma de Protozoos , Genotipo , Humanos , Filogenia , Plasmodium falciparum/clasificación , Polimorfismo de Nucleótido Simple , Análisis de Componente Principal
18.
Pharmacol Res ; 117: 192-217, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27867026

RESUMEN

Parasitic protozoan diseases continue to rank among the world's greatest global health problems, which are also common among poor populations. Currently available drugs for treatment present drawbacks, urging the need for more effective, safer, and cheaper drugs. Artemisinin (ART) and its derivatives are some of the most important classes of antimalarial agents originally derived from Artemisia annua L. However, besides the outstanding antimalarial and antischistosomal activities, ART and its derivatives also possess activities against other parasitic protozoa. In this paper we review the activities of ART and its derivatives against protozoan parasites in vitro and in vivo, including Leishmania spp., Trypanosoma spp., Toxoplasma gondii, Neospora caninum, Eimeria tenella, Acanthamoeba castellanii, Naegleria fowleri, Cryptosporidium parvum, Giardia lamblia, and Babesia spp. We conclude that ART and its derivatives may be good alternatives for treating other non-malarial protozoan infections in developing countries, although more studies are necessary before they can be applied clinically.


Asunto(s)
Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Artemisininas/farmacología , Artemisininas/uso terapéutico , Malaria/tratamiento farmacológico , Infecciones por Protozoos/tratamiento farmacológico , Animales , Humanos
19.
Proc Natl Acad Sci U S A ; 111(4): E511-20, 2014 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-24474800

RESUMEN

Malaria infection triggers vigorous host immune responses; however, the parasite ligands, host receptors, and the signaling pathways responsible for these reactions remain unknown or controversial. Malaria parasites primarily reside within RBCs, thereby hiding themselves from direct contact and recognition by host immune cells. Host responses to malaria infection are very different from those elicited by bacterial and viral infections and the host receptors recognizing parasite ligands have been elusive. Here we investigated mouse genome-wide transcriptional responses to infections with two strains of Plasmodium yoelii (N67 and N67C) and discovered differences in innate response pathways corresponding to strain-specific disease phenotypes. Using in vitro RNAi-based gene knockdown and KO mice, we demonstrated that a strong type I IFN (IFN-I) response triggered by RNA polymerase III and melanoma differentiation-associated protein 5, not Toll-like receptors (TLRs), binding of parasite DNA/RNA contributed to a decline of parasitemia in N67-infected mice. We showed that conventional dendritic cells were the major sources of early IFN-I, and that surface expression of phosphatidylserine on infected RBCs might promote their phagocytic uptake, leading to the release of parasite ligands and the IFN-I response in N67 infection. In contrast, an elevated inflammatory response mediated by CD14/TLR and p38 signaling played a role in disease severity and early host death in N67C-infected mice. In addition to identifying cytosolic DNA/RNA sensors and signaling pathways previously unrecognized in malaria infection, our study demonstrates the importance of parasite genetic backgrounds in malaria pathology and provides important information for studying human malaria pathogenesis.


Asunto(s)
Interacciones Huésped-Parásitos , Inmunidad Innata , Malaria/inmunología , Parasitemia/inmunología , Plasmodium yoelii/fisiología , Transducción de Señal , Anciano , Animales , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Interferón Tipo I/metabolismo , Malaria/mortalidad , Malaria/parasitología , Ratones , Ratones Noqueados , Parasitemia/parasitología , Fagocitosis , Plasmodium yoelii/inmunología
20.
Malar J ; 15: 30, 2016 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-26791272

RESUMEN

BACKGROUND: The rodent malaria parasite Plasmodium yoelii is an important animal model for studying host-parasite interaction and molecular basis of malaria pathogenesis. Although a draft genome of P. yoelii yoelii YM is available, and RNA sequencing (RNA-seq) data for several rodent malaria species (RMP) were reported recently, variations in coding regions and structure of mRNA transcript are likely present between different parasite strains or subspecies. Sequencing of cDNA libraries from additional parasite strains/subspecies will help improve the gene models and genome annotation. METHODS: Here two directional cDNA libraries from mixed blood stages of a subspecies of P. yoelii (P. y. nigeriensis NSM) with or without mefloquine (MQ) treatment were sequenced, and the sequence reads were compared to the genome and cDNA sequences of P. y. yoelii YM in public databases to investigate single nucleotide polymorphisms (SNPs) in coding regions, variations in intron-exon structure and differential splicing between P. yoelii subspecies, and variations in gene expression under MQ pressure. RESULTS: Approximately 56 million of 100 bp paired-end reads were obtained, providing an average of ~225-fold coverage for the coding regions. Comparison of the sequence reads to the YM genome revealed introns in 5' and 3' untranslated regions (UTRs), altered intron/exon boundaries, alternative splicing, overlapping sense-antisense reads, and potentially new transcripts. Interestingly, comparison of the NSM RNA-seq reads obtained here with those of YM discovered differentially spliced introns; e.g., spliced introns in one subspecies but not the other. Alignment of the NSM cDNA sequences to the YM genome sequence also identified ~84,000 SNPs between the two parasites. CONCLUSION: The discoveries of UTR introns and differentially spliced introns between P. yoelii subspecies raise interesting questions on the potential role of these introns in regulating gene expression and evolution of malaria parasites.


Asunto(s)
Empalme Alternativo/genética , Intrones/genética , Plasmodium yoelii/genética , ARN sin Sentido/genética , Genoma de Protozoos/genética , Malaria/parasitología , Polimorfismo de Nucleótido Simple/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA