Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Front Cell Infect Microbiol ; 14: 1332381, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38357442

RESUMEN

Visceral leishmaniasis (VL) is the most severe type of leishmaniasis which is caused by infection of Leishmania donovani complex. In the BALB/c mouse model of VL, multinucleated giant cells (MGCs) with heavy parasite infection consist of the largest population of hemophagocytes in the spleen of L. donovani-infected mice, indicating that MGCs provide the parasites a circumstance beneficial for their survival. Although ATP6V0D2 is a demonstrated factor inducing the formation of hemophagocytic MGCs during L. donovani infection, functions of this protein in shaping the infection outcome in macrophages remain unclear. Here we evaluated the influence of upregulated ATP6V0D2 on intracellular survival of the parasites. L. donovani infection-induced hemophagocytosis of normal erythrocytes by macrophages was suppressed by RNAi-based knockdown of Atp6v0d2. The knockdown of Atp6v0d2 did not improve the survival of amastigotes within macrophages when the cells were cultured in the absence of erythrocytes. On the other hand, reduced intracellular survival of amastigotes in macrophages by the knockdown was observed when macrophages were supplemented with antibody-opsonized erythrocytes before infection. There, increase in cytosolic labile iron pool was observed in the L. donovani-infected knocked-down macrophages. It suggests that ATP6V0D2 plays roles not only in upregulation of hemophagocytosis but also in iron trafficking within L. donovani-infected macrophages. Superior access to iron in macrophages may be how the upregulated expression of the molecule brings benefit to Leishmania for their intracellular survival in the presence of erythrocytes.


Asunto(s)
Leishmania donovani , Leishmaniasis Visceral , Animales , Ratones , Eritrocitos , Hierro/metabolismo , Leishmaniasis Visceral/parasitología , Macrófagos/metabolismo , Ratones Endogámicos BALB C , Regulación hacia Arriba
2.
Viruses ; 15(6)2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-37376669

RESUMEN

Bats are of significant interest as reservoirs for various zoonotic viruses with high diversity. During the past two decades, many herpesviruses have been identified in various bats worldwide by genetic approaches, whereas there have been few reports on the isolation of infectious herpesviruses. Herein, we report the prevalence of herpesvirus infection of bats captured in Zambia and genetic characterization of novel gammaherpesviruses isolated from striped leaf-nosed bats (Macronycteris vittatus). By our PCR screening, herpesvirus DNA polymerase (DPOL) genes were detected in 29.2% (7/24) of Egyptian fruit bats (Rousettus aegyptiacus), 78.1% (82/105) of Macronycteris vittatus, and one Sundevall's roundleaf bat (Hipposideros caffer) in Zambia. Phylogenetic analyses of the detected partial DPOL genes revealed that the Zambian bat herpesviruses were divided into seven betaherpesvirus groups and five gammaherpesvirus groups. Two infectious strains of a novel gammaherpesvirus, tentatively named Macronycteris gammaherpesvirus 1 (MaGHV1), were successfully isolated from Macronycteris vittatus bats, and their complete genomes were sequenced. The genome of MaGHV1 encoded 79 open reading frames, and phylogenic analyses of the DNA polymerase and glycoprotein B demonstrated that MaGHV1 formed an independent lineage sharing a common origin with other bat-derived gammaherpesviruses. Our findings provide new information regarding the genetic diversity of herpesviruses maintained in African bats.


Asunto(s)
Quirópteros , Gammaherpesvirinae , Herpesviridae , Animales , Filogenia , Zambia/epidemiología , Herpesviridae/genética
3.
Front Cell Infect Microbiol ; 12: 953785, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36211967

RESUMEN

Leishmaniasis is caused by infection with protozoan parasites of the genus Leishmania. In both clinical and experimental visceral leishmaniasis, macrophage multinucleation is observed in parasitized tissues. However, the feature and the mechanism of macrophage multinucleation remained unclear. Here, we report that infection of Leishmania donovani, a causative agent of visceral leishmaniasis, induces multinucleation of bone marrow-derived macrophages (BMDMs) in vitro. When these infection-induced multinucleated macrophages were compared with cytokine-induced multinucleated giant cells, the former had higher phagocytic activity on red blood cells but no apparent changes on phagocytosis of latex beads. BMDMs infected with L. donovani had increased expression of ATP6V0D2, one of the components of V-ATPase, which was also upregulated in the spleen of infected mice. Infection-induced ATP6V0D2 localized in a cytoplasmic compartment, which did not overlap with the mitochondria, endoplasmic reticulum, or lysosomes. When ATP6V0D2 expression was recombinantly induced in BMDMs, the formation of multinucleated macrophages was induced as seen in the infected macrophages. Taken together, L. donovani infection induces multinucleation of macrophages via ATP6V0D2 upregulation leading to a unique metamorphosis of the macrophages toward hemophagocytes.


Asunto(s)
Leishmania donovani , Leishmaniasis Visceral , ATPasas de Translocación de Protón Vacuolares , Adenosina Trifosfatasas , Animales , Citocinas , Células Gigantes , Leishmaniasis Visceral/parasitología , Ratones , Ratones Endogámicos BALB C , Regulación hacia Arriba
4.
J Neuroinflammation ; 19(1): 13, 2022 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-35012562

RESUMEN

BACKGROUND: Postviral olfactory dysfunction (PVOD) following a viral upper respiratory tract infection (URI) is one of the most common causes of olfactory disorders, often lasting for over a year. To date, the molecular pathology of PVOD has not been elucidated. METHODS: A murine model of Toll-like receptor 3 (TLR3)-mediated upper respiratory tract inflammation was used to investigate the impact of URIs on the olfactory system. Inflammation was induced via the intranasal administration of polyinosinic-polycytidylic acid (poly(I:C), a TLR3 ligand) to the right nostril for 3 days. Peripheral olfactory sensory neurons (OSNs), immune cells in the olfactory mucosa, and glial cells in the olfactory bulb (OB) were analyzed histologically. Proinflammatory cytokines in the nasal tissue and OB were evaluated using the quantitative real-time polymerase chain reaction (qPCR) and enzyme-linked immunosorbent assay (ELISA). RESULTS: In the treated mice, OSNs were markedly reduced in the olfactory mucosa, and T cell and neutrophil infiltration therein was observed 1 day after the end of poly(I:C) administration. Moreover, there was a considerable increase in microglial cells and slight increase in activated astrocytes in the OB. In addition, qPCR and ELISA revealed the elevated expression of interleukin-1 beta, interleukin-6, tumor necrosis factor-alpha, and interferon-gamma both in the OB and nasal tissue. CONCLUSIONS: Taken together, the decreased peripheral OSNs, OB microgliosis, and elevated proinflammatory cytokines suggest that immunological changes in the OB may be involved in the pathogenesis of PVOD.


Asunto(s)
Inflamación/metabolismo , Bulbo Olfatorio/metabolismo , Infecciones del Sistema Respiratorio/metabolismo , Receptor Toll-Like 3/metabolismo , Animales , Modelos Animales de Enfermedad , Inflamación/inducido químicamente , Ratones , Microglía/metabolismo , Mucosa Olfatoria/metabolismo , Poli I-C/farmacología
5.
Front Vet Sci ; 8: 599815, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33585616

RESUMEN

We clarified the genetic diversity of Trypanosoma spp. within the Kafue ecosystem, using PCR targeting the internal transcribed spacer 1 and the cathepsin L-like cysteine protease (CatL) sequences. The overall prevalence of Trypanosoma spp. in cattle and tsetse flies was 12.65 and 26.85%, respectively. Cattle positive for Trypanosoma vivax had a significantly lower packed cell volume, suggesting that T. vivax is the dominant Trypanosoma spp. causing anemia in this area. Among the 12 operational taxonomic units (OTUs) of T. vivax CatL sequences detected, one was from a known T. vivax lineage, two OTUs were from known T. vivax-like lineages, and nine OTUs were considered novel T. vivax-like lineages. These findings support previous reports that indicated the extensive diversity of T. vivax-like lineages. The findings also indicate that combining CatL PCR with next generation sequencing is useful in assessing Trypanosoma spp. diversity, especially for T. vivax and T. vivax-like lineages. In addition, the 5.42% prevalence of Trypanosoma brucei rhodesiense found in cattle raises concern in the community and requires careful monitoring of human African trypanosomiasis.

6.
PLoS Negl Trop Dis ; 13(11): e0007816, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31738750

RESUMEN

Visceral leishmaniasis (VL) is caused by parasitic protozoa of the genus Leishmania and is characterized by clinical manifestations such as fever, hepatosplenomegaly and anemia. Hemophagocytosis, the phenomenon of phagocytosis of blood cells by macrophages, is found in VL patients. In a previous study we established an experimental model of VL, reproducing anemia in mice for the first time, and identified hemophagocytosis by heavily infected macrophages in the spleen as a possible cause of anemia. However, the mechanism for parasite-induced hemophagocytosis or its role in parasite survival remained unclear. Here, we established an in vitro model of Leishmania-induced hemophagocytosis to explore the molecules involved in this process. In contrast to naïve RAW264.7 cells (mouse macrophage cell line) which did not uptake freshly isolated erythrocytes, RAW264.7 cells infected with L. donovani showed enhanced phagocytosis of erythrocytes. Additionally, for hemophagocytes found both in vitro and in vivo, the expression of signal regulatory protein α (SIRPα), one of the receptors responsible for the 'don't-eat-me' signal was suppressed by post-transcriptional control. Furthermore, the overlapped phagocytosis of erythrocytes and Leishmania parasites within a given macrophage appeared to be beneficial to the parasites; the in vitro experiments showed a higher number of parasites within macrophages that had been induced to engulf erythrocytes. Together, these results suggest that Leishmania parasites may actively induce hemophagocytosis by manipulating the expression of SIRPα in macrophages/hemophagocytes, in order to secure their parasitism.


Asunto(s)
Leishmania donovani/fisiología , Linfohistiocitosis Hemofagocítica , Macrófagos/parasitología , Fagocitosis , Animales , Línea Celular , Modelos Animales de Enfermedad , Eritrocitos , Leishmania donovani/patogenicidad , Leishmaniasis Visceral/parasitología , Masculino , Ratones , Ratones Endogámicos BALB C , Células RAW 264.7 , Bazo/parasitología , Transcriptoma
7.
Cell Microbiol ; 20(9): e12852, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29691989

RESUMEN

The intracellular pathogen Legionella pneumophila influences numerous eukaryotic cellular processes through the Dot/Icm-dependent translocation of more than 300 effector proteins into the host cell. Although many translocated effectors localise to the Legionella replicative vacuole, other effectors can affect remote intracellular sites. Following infection, a subset of effector proteins localises to the nucleus where they subvert host cell transcriptional responses to infection. Here, we identified Lpw27461 (Lpp2587), Lpg2519 as a new nuclear-localised effector that we have termed SnpL. Upon ectopic expression or during L. pneumophila infection, SnpL showed strong nuclear localisation by immunofluorescence microscopy but was excluded from nucleoli. Using immunoprecipitation and mass spectrometry, we determined the host-binding partner of SnpL as the eukaryotic transcription elongation factor, Suppressor of Ty5 (SUPT5H)/Spt5. SUPT5H is an evolutionarily conserved component of the DRB sensitivity-inducing factor complex that regulates RNA Polymerase II dependent mRNA processing and transcription elongation. Protein interaction studies showed that SnpL bound to the central Kyprides, Ouzounis, Woese motif region of SUPT5H. Ectopic expression of SnpL led to massive upregulation of host gene expression and macrophage cell death. The activity of SnpL further highlights the ability of L. pneumophila to control fundamental eukaryotic processes such as transcription that, in the case of SnpL, leads to global upregulation of host gene expression.


Asunto(s)
Interacciones Huésped-Patógeno , Legionella pneumophila/patogenicidad , Proteínas de Transporte de Membrana/metabolismo , Proteínas Nucleares/metabolismo , ARN Polimerasa II/metabolismo , Factores de Elongación Transcripcional/metabolismo , Factores de Virulencia/metabolismo , Animales , Muerte Celular , Línea Celular , Núcleo Celular/química , Humanos , Inmunoprecipitación , Macrófagos/microbiología , Macrófagos/fisiología , Espectrometría de Masas , Microscopía Fluorescente , Unión Proteica , Transporte de Proteínas
8.
Cell Rep ; 19(9): 1874-1887, 2017 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-28564605

RESUMEN

Cancer patients having anti-programmed cell death-1 (PD-1)/PD ligand 1 (L1)-unresponsive tumors may benefit from advanced immunotherapy. Double-stranded RNA triggers dendritic cell (DC) maturation to cross-prime antigen-specific cytotoxic T lymphocytes (CTLs) via Toll-like receptor 3 (TLR3). The TLR3-specific RNA agonist, ARNAX, can induce anti-tumor CTLs without systemic cytokine/interferon (IFN) production. Here, we have developed a safe vaccine adjuvant for cancer that effectively implements anti-PD-L1 therapy. Co-administration of ARNAX with a tumor-associated antigen facilitated tumor regression in mouse models, and in combination with anti-PD-L1 antibody, activated tumor-specific CTLs in lymphoid tissues, enhanced CTL infiltration, and overcame anti-PD-1 resistance without cytokinemia. The TLR3-TICAM-1-interferon regulatory factor (IRF)3-IFN-ß axis in DCs exclusively participated in CD8+ T cell cross-priming. ARNAX therapy established Th1 immunity in the tumor microenvironment, upregulating genes involved in DC/T cell/natural killer (NK) cell recruitment and functionality. Human ex vivo studies disclosed that ARNAX+antigen induced antigen-specific CTL priming and proliferation in peripheral blood mononuclear cells (PBMCs), supporting the feasibility of ARNAX for potentiating anti-PD-1/PD-L1 therapy in human vaccine immunotherapy.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Antígeno B7-H1/metabolismo , Vacunas contra el Cáncer/inmunología , Citocinas/toxicidad , Inmunidad Innata , Inmunoterapia , Receptor Toll-Like 3/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Anticuerpos Antineoplásicos/farmacología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Femenino , Humanos , Inmunidad Innata/efectos de los fármacos , Terapia de Inmunosupresión , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/metabolismo , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Transducción de Señal/efectos de los fármacos , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Células TH1/efectos de los fármacos , Células TH1/inmunología , Microambiente Tumoral/efectos de los fármacos
9.
Sci Rep ; 6: 34349, 2016 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-27686861

RESUMEN

CRK and CRKL adapter proteins play essential roles in development and cancer through their SRC homology 2 and 3 (SH2 and SH3) domains. To gain insight into the origin of their shared functions, we have investigated their evolutionary history. We propose a term, crk/crkl ancestral (crka), for orthologs in invertebrates before the divergence of CRK and CRKL in the vertebrate ancestor. We have isolated two orthologs expressed in the choanoflagellate Monosiga brevicollis, a unicellular relative to the metazoans. Consistent with its highly-conserved three-dimensional structure, the SH2 domain of M. brevicollis crka1 can bind to the mammalian CRK/CRKL SH2 binding consensus phospho-YxxP, and to the SRC substrate/focal adhesion protein BCAR1 (p130CAS) in the presence of activated SRC. These results demonstrate an ancient origin of the CRK/CRKL SH2-target recognition specificity. Although BCAR1 orthologs exist only in metazoans as identified by an N-terminal SH3 domain, YxxP motifs, and a C-terminal FAT-like domain, some pre-metazoan transmembrane proteins include several YxxP repeats in their cytosolic region, suggesting that they are remotely related to the BCAR1 substrate domain. Since the tyrosine kinase SRC also has a pre-metazoan origin, co-option of BCAR1-related sequences may have rewired the crka-dependent network to mediate adhesion signals in the metazoan ancestor.

10.
Parasitol Res ; 115(8): 3139-45, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27095567

RESUMEN

Cysteine-based peroxidases, known as peroxiredoxins (Prx) or thioredoxin peroxidases (TPx), are important antioxidant enzymes that prevent oxidative damage caused by reactive oxygen species (ROS). In this study, we identified a novel mitochondrial 2-Cys Prx, BbTPx-2, from a bovine Babesia parasite, B. bovis. BbTPx-2 complementary DNA (cDNA) encodes a polypeptide of 254 amino acid residues. This protein has a mitochondrial targeting peptide at the N-terminus and two conserved cysteine residues of the typical 2-Cys Prx. By using a thiol mixed-function oxidation assay, the antioxidant activity of recombinant BbTPx-2 was revealed, and its antioxidant activity was comparable to that of a cytosolic 2-Cys Prx from B. bovis, BbTPx-1. Notably, we confirmed that BbTPx-2 was expressed in the mitochondrion of B. bovis merozoites. Taken together, the results suggest that the mitochondrial BbTPx-2 is an antioxidative enzyme for scavenging ROS in B. bovis.


Asunto(s)
Antioxidantes/metabolismo , Babesia bovis/enzimología , Mitocondrias/enzimología , Peroxirredoxinas/metabolismo , Secuencia de Aminoácidos , Animales , Babesia bovis/metabolismo , Secuencia de Bases , Bovinos , Cisteína/química , ADN Complementario/genética , Mitocondrias/genética , Oxidación-Reducción , Peroxirredoxinas/genética , Especies Reactivas de Oxígeno/metabolismo , Alineación de Secuencia
11.
Parasit Vectors ; 8: 654, 2015 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-26694921

RESUMEN

BACKGROUND: The glyoxalase pathway, which includes two enzymes, glyoxalase 1 and 2 (Glo1 and Glo2), is a ubiquitous cellular system responsible for the removal of cytotoxic methylglyoxal produced during glycolysis. Protozoan parasites, including Toxoplasma gondii (T. gondii) tachyzoites, produce methylglyoxal because of increased glycolytic fluxes. A Glo1 inhibitor such as curcumin could be considered a drug candidate for anti-protozoan, anti-inflammatory, and anti-cancer therapy. METHODS: The T. gondii Glo1 gene (TgGlo1) was cloned and the recombinant protein was produced. Enzyme kinetics of TgGlo1 and five mutants were evaluated by adding methylglyoxal and glutathione to a reaction mixture. Finally, the inhibitory effects of various concentrations of curcumin on recombinant TgGlo1 were evaluated using in vitro cultures of T. gondii. RESULTS: Active recombinant TgGlo1 was successfully produced and the active sites (E166 and E251) of TgGlo1 were verified by point mutagenesis. Curcumin at the tested doses inhibited the enzymatic activity of recombinant TgGlo1 as well as the parasitic propagation of in vitro-cultured T. gondii. The Ki and IC50 were 12.9 ± 0.5 µM and 38.3 ± 0.9 µM, respectively. CONCLUSION: The inhibitory effect of curcumin on the enzymatic activity of TgGlo1 and parasitic propagation of T. gondii could be explored in the potential development of a potent drug for the treatment of toxoplasmosis. However, considering the fact that curcumin is known to have many effects on other molecules in the micromolar range, further elucidation of curcumin's direct inhibition of the glyoxalase system of T. gondii will be needed.


Asunto(s)
Antiprotozoarios/metabolismo , Curcumina/metabolismo , Inhibidores Enzimáticos/metabolismo , Lactoilglutatión Liasa/genética , Lactoilglutatión Liasa/metabolismo , Toxoplasma/efectos de los fármacos , Toxoplasma/enzimología , Animales , Dominio Catalítico , Clonación Molecular , Análisis Mutacional de ADN , Expresión Génica , Glutatión/metabolismo , Cinética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación Puntual , Piruvaldehído/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Toxoplasma/genética
12.
J Comput Chem ; 35(29): 2132-9, 2014 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-25220475

RESUMEN

The Poisson-Boltzmann implicit solvent (PB) is widely used to estimate the solvation free energies of biomolecules in molecular simulations. An optimized set of atomic radii (PB radii) is an important parameter for PB calculations, which determines the distribution of dielectric constants around the solute. We here present new PB radii for the AMBER protein force field to accurately reproduce the solvation free energies obtained from explicit solvent simulations. The presented PB radii were optimized using results from explicit solvent simulations of the large systems. In addition, we discriminated PB radii for N- and C-terminal residues from those for nonterminal residues. The performances using our PB radii showed high accuracy for the estimation of solvation free energies at the level of the molecular fragment. The obtained PB radii are effective for the detailed analysis of the solvation effects of biomolecules.


Asunto(s)
Simulación de Dinámica Molecular , Péptidos/química , Proteínas/química , Termodinámica , Solubilidad , Solventes/química
13.
Genome Res ; 24(9): 1433-44, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25091627

RESUMEN

To understand the molecular mechanisms of parasitism in vivo, it is essential to elucidate how the transcriptomes of the human hosts and the infecting parasites affect one another. Here we report the RNA-seq analysis of 116 Indonesian patients infected with the malaria parasite Plasmodium falciparum (Pf). We extracted RNAs from their peripheral blood as a mixture of host and parasite transcripts and mapped the RNA-seq tags to the human and Pf reference genomes to separate the respective tags. We were thus able to simultaneously analyze expression patterns in both humans and parasites. We identified human and parasite genes and pathways that correlated with various clinical data, which may serve as primary targets for drug developments. Of particular importance, we revealed characteristic expression changes in the human innate immune response pathway genes including TLR2 and TICAM2 that correlated with the severity of the malaria infection. We also found a group of transcription regulatory factors, JUND, for example, and signaling molecules, TNFAIP3, for example, that were strongly correlated in the expression patterns of humans and parasites. We also identified several genetic variations in important anti-malaria drug resistance-related genes. Furthermore, we identified the genetic variations which are potentially associated with severe malaria symptoms both in humans and parasites. The newly generated data should collectively lay a unique foundation for understanding variable behaviors of the field malaria parasites, which are far more complex than those observed under laboratory conditions.


Asunto(s)
Genoma Humano , Genoma de Protozoos , Malaria/genética , Plasmodium falciparum/genética , Transcriptoma , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adolescente , Adulto , Antimaláricos/uso terapéutico , Estudios de Casos y Controles , Niño , Preescolar , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Resistencia a Medicamentos/genética , Etiquetas de Secuencia Expresada , Femenino , Interacciones Huésped-Parásitos/genética , Humanos , Inmunidad Innata/genética , Lactante , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Malaria/diagnóstico , Malaria/tratamiento farmacológico , Masculino , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Plasmodium falciparum/patogenicidad , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Virulencia/genética
14.
Mol Biochem Parasitol ; 191(1): 36-43, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24041588

RESUMEN

It is known that gene expression in kinetoplastida is regulated post-transcriptionally. Several previous studies have shown that stage-specific gene expression in trypanosomes is regulated by cis-elements located in the 3' untranslated region (UTR) of each mRNA and also by RNA binding proteins. Our previous study revealed that gene expression of congolense epimastigote specific protein (cesp) was regulated by cis-elements located in the 3'UTR. In the present study, we identified the adenosine and uridine rich region in the cesp 3'UTR. Using transgenic trypanosome cell lines with different egfp expression cassettes, we showed that this adenosine and uridine rich region is one of the regulatory elements for epimastigote form (EMF) stage-specific gene expression via the regulatory cis-element of the eukaryotic AU rich element (ARE). Therefore this required element within the cesp 3'UTR was designated as T. congolense ARE. This required cis-element might selectively stabilize mRNA in the EMF stage and destabilize mRNA in other stages. By RNA electro mobility shift assay, unknown stage-specific RNA binding proteins (RBPs) whose sequences specifically interacted with the required cis-element were found. These results indicate that EMF stage specific cis-element and RBP complexes might specifically stabilize cesp mRNA in EMF.


Asunto(s)
Proteínas Protozoarias/biosíntesis , Proteínas Protozoarias/genética , Trypanosoma congolense/crecimiento & desarrollo , Trypanosoma congolense/genética , Regiones no Traducidas 3' , Fusión Artificial Génica , Análisis Mutacional de ADN , Ensayo de Cambio de Movilidad Electroforética , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Organismos Modificados Genéticamente , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Protozoario/genética , ARN Protozoario/metabolismo , Proteínas de Unión al ARN/metabolismo
15.
Parasitol Int ; 61(3): 481-6, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22537971

RESUMEN

A heterologous prime-boost strategy with priming plasmid DNA followed by recombinant virus expressing relevant antigens is known to stimulate protective immunity against intracellular parasites. In this study, we have evaluated a heterologous prime-boost strategy for immunizing mice against Toxoplasma gondii infection. Our results revealed that the prime-boost strategy using both plasmid DNA and adenoviral vector encoding TgAMA1 may stimulate both humoral and Th1/Th2 cellular immune responses specific for TgAMA1. Moreover, C57BL/6 mice immunized with the pAMA1/Ad5Null, pNull/Ad5AMA1, and pAMA1/Ad5AMA1 constructs showed survival rates of 12.5%, 37.5%, and 50%, respectively. In contrast, all the pNull/Ad5Null immunized mice died after infection with the PLK-GFP strain of T. gondii. Brain cyst burden was reduced by 23% in mice immunized with pAMA1/Ad5AMA1 compared with the pNull/Ad5AMA1 immunized mice. These results demonstrate that the heterologous DNA priming and recombinant adenovirus boost strategy may provide protective immunity against T. gondii infection.


Asunto(s)
Antígenos de Protozoos/inmunología , Vacunas Antiprotozoos/genética , Toxoplasmosis/inmunología , Toxoplasmosis/prevención & control , Vacunación/métodos , Adenoviridae/genética , Animales , Antígenos de Protozoos/genética , Antígenos de Protozoos/metabolismo , ADN Protozoario/genética , ADN Protozoario/aislamiento & purificación , Femenino , Regulación de la Expresión Génica , Vectores Genéticos , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Plásmidos , Vacunas Antiprotozoos/inmunología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Balance Th1 - Th2 , Toxoplasma/genética , Toxoplasma/crecimiento & desarrollo , Toxoplasma/inmunología
16.
Infect Immun ; 80(1): 311-20, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22064713

RESUMEN

Although primary infection of mice with Babesia microti has been shown to protect mice against subsequent lethal infection by Babesia rodhaini, the mechanism behind the cross-protection is unknown. To unravel this mechanism, we investigated the influence of primary infection of mice with nonlethal B. microti using different time courses on the outcome of subsequent lethal B. rodhaini infection. Simultaneous infections of mice with these parasites resulted in rapid increases in parasitemia, with 100% mortality in BALB/c mice, as observed with control mice infected with B. rodhaini alone. In contrast, mice with acute, resolving, and chronic-phase B. microti infections were completely protected against B. rodhaini, resulting in low parasitemia and no mortalities. Mice immunized with dead B. microti were not protected from B. rodhaini infection, although high antibody responses were induced. Interestingly, the protected mice had significantly decreased levels of antibody response, cytokines (including gamma interferon [IFN-γ], interleukin-2 [IL-2], IL-8, IL-10, and IL-12), and nitric oxide levels after infection with B. rodhaini. SCID mice and IFN-γ-deficient mice with chronic B. microti infections demonstrated protective responses comparable to those of immunocompetent mice. Likewise, in vivo NK cell depletion did not significantly impair the protective responses. Conversely, macrophage depletion resulted in increased susceptibility to B. rodhaini infection associated with changes in their antibody and cytokines profiles, indicating that macrophages contribute to the protection against this challenge infection. We conclude that future development of vaccines against Babesia should include a strategy that enhances the appropriate activation of macrophages.


Asunto(s)
Babesia/inmunología , Babesiosis/inmunología , Babesiosis/parasitología , Protección Cruzada , Macrófagos/inmunología , Macrófagos/parasitología , Animales , Anticuerpos Antiprotozoarios/sangre , Babesiosis/mortalidad , Babesiosis/prevención & control , Citocinas/metabolismo , Femenino , Procedimientos de Reducción del Leucocitos , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Parasitemia/inmunología , Parasitemia/mortalidad , Parasitemia/parasitología , Parasitemia/prevención & control , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA