Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Cell Infect Microbiol ; 11: 772311, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34858879

RESUMEN

Until 2015, loss-of-function studies to elucidate protein function in Leishmania relied on gene disruption through homologous recombination. Then, the CRISPR/Cas9 revolution reached these protozoan parasites allowing efficient genome editing with one round of transfection. In addition, the development of LeishGEdit, a PCR-based toolkit for generating knockouts and tagged lines using CRISPR/Cas9, allowed a more straightforward and effective genome editing. In this system, the plasmid pTB007 is delivered to Leishmania for episomal expression or integration in the ß-tubulin locus and for the stable expression of T7 RNA polymerase and Cas9. In South America, and especially in Brazil, Leishmania (Viannia) braziliensis is the most frequent etiological agent of tegumentary leishmaniasis. The L. braziliensis ß-tubulin locus presents significant sequence divergence in comparison with Leishmania major, which precludes the efficient integration of pTB007 and the stable expression of Cas9. To overcome this limitation, the L. major ß-tubulin sequences, present in the pTB007, were replaced by a Leishmania (Viannia) ß-tubulin conserved sequence generating the pTB007_Viannia plasmid. This modification allowed the successful integration of the pTB007_Viannia cassette in the L. braziliensis M2903 genome, and in silico predictions suggest that this can also be achieved in other Viannia species. The activity of Cas9 was evaluated by knocking out the flagellar protein PF16, which caused a phenotype of immobility in these transfectants. Endogenous PF16 was also successfully tagged with mNeonGreen, and an in-locus complementation strategy was employed to return a C-terminally tagged copy of the PF16 gene to the original locus, which resulted in the recovery of swimming capacity. The modified plasmid pTB007_Viannia allowed the integration and stable expression of both T7 RNA polymerase and Cas9 in L. braziliensis and provided an important tool for the study of the biology of this parasite.


Asunto(s)
Leishmania braziliensis , Leishmania major , Sistemas CRISPR-Cas , ARN Polimerasas Dirigidas por ADN , Edición Génica , Leishmania braziliensis/genética , Proteínas Virales
2.
Dev Dyn ; 250(2): 249-262, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32562595

RESUMEN

BACKGROUND: During embryonic development, complex changes in cell behavior generate the final form of the tissues. Extension of cell protrusions have been described as an important component in this process. Cellular protrusions have been associated with generation of traction, intercellular communication or establishment of signaling gradients. Here, we describe and compare in detail from live imaging data the dynamics of protrusions in the surface ectoderm of chick and mouse embryos. In particular, we explore the differences between cells surrounding the lens placode and other regions of the head. RESULTS: Our results showed that protrusions from the eye region in mouse embryos are longer than those in chick embryos. In addition, protrusions from regions where there are no significant changes in tissue shape are longer and more stable than protrusions that surround the invaginating lens placode. We did not find a clear directionality to the protrusions in any region. Finally, we observed intercellular trafficking of membrane puncta in the protrusions of both embryos in all the regions analyzed. CONCLUSIONS: In summary, the results presented here suggest that the dynamics of these protrusions adapt to their surroundings and possibly contribute to intercellular communication in embryonic cephalic epithelia.


Asunto(s)
Extensiones de la Superficie Celular , Ectodermo/citología , Animales , Embrión de Pollo , Ratones , Morfogénesis
3.
Cell Biochem Biophys ; 78(3): 391-398, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32681442

RESUMEN

Impaired periodontal healing is a common complication of diabetes mellitus (DM), frequently related to hyperglycemia. MicroRNAs 221 and 222 have been studied as biomarkers for inflammatory diseases, including diabetes, but their role in the periodontal ligament (PL) is unknown. The effects of high glucose on human PL cells death were studied, as well as the expression of microRNA-221 and microRNA-222, potentially modulated by DM. Cells were obtained from the premolar teeth of young humans and cultured for 7 days under different glucose concentrations (5 or 30 mM). MicroRNAs-221/222 expressions were evaluated by real-time RT-PCR and apoptosis by TUNEL assays. Caspase-3 expression was studied by western blotting and immunocytochemistry. High glucose increased apoptosis and caspase-3 protein expression by about 3×. MicroRNA-221 and microRNA-222 expressions decreased by nearly 40% under high glucose. MicroRNA-221 and microRNA-222 inhibition using antagomiRs increased apoptosis by 2-3×, while the expression of caspase-3, a validated target for these microRNAs, was increased by 50%. The overexpression of both microRNAs using miR mimics in high glucose cells did no effect on apoptosis but increased caspase-3 expression by 30%. In conclusion, high glucose induces apoptosis of human PL cells potentially through a reduction of microRNA-221 and microRNA-222 expression and elevation of caspase-3.


Asunto(s)
Apoptosis , Glucosa/metabolismo , MicroARNs/genética , Ligamento Periodontal/citología , Adolescente , Diente Premolar/citología , Caspasa 3/metabolismo , Células Cultivadas , Niño , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección
4.
Gene Ther ; 27(1-2): 51-61, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31439890

RESUMEN

Cancer therapies that target a single protein or pathway may be limited by their specificity, thus missing key players that control cellular proliferation and contributing to the failure of the treatment. We propose that approaches to cancer therapy that hit multiple targets would limit the chances of escape. To this end, we have developed a bicistronic adenoviral vector encoding both the CDKN2A and p53 tumor suppressor genes. The bicistronic vector, AdCDKN2A-I-p53, supports the translation of both gene products from a single transcript, assuring that all transduced cells will express both proteins. We show that combined, but not single, gene transfer results in markedly reduced proliferation and increased cell death correlated with reduced levels of phosphorylated pRB, induction of CDKN1A and caspase 3 activity, yet avoiding the induction of senescence. Using isogenic cell lines, we show that these effects were not impeded by the presence of mutant p53. In a mouse model of in situ gene therapy, a single intratumoral treatment with the bicistronic vector conferred markedly inhibited tumor progression while the treatment with either CDKN2A or p53 alone only partially controlled tumor growth. Histologic analysis revealed widespread transduction, yet reduced proliferation and increased cell death was associated only with the simultaneous transfer of CDKN2A and p53. We propose that restoration of two of the most frequently altered genes in human cancer, mediated by AdCDKN2A-I-p53, is beneficial since multiple targets are reached, thus increasing the efficacy of the treatment.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/administración & dosificación , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Terapia Genética/métodos , Neoplasias Pulmonares/terapia , Proteína p53 Supresora de Tumor/administración & dosificación , Proteína p53 Supresora de Tumor/genética , Adenoviridae/genética , Animales , Apoptosis/fisiología , Línea Celular Tumoral , Proliferación Celular/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Femenino , Genes p53 , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Células HEK293 , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Transducción Genética/métodos , Proteína p53 Supresora de Tumor/biosíntesis , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Int J Parasitol Drugs Drug Resist ; 11: 139-147, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30850347

RESUMEN

In Brazil, cutaneous leishmaniasis is caused predominantly by L. (V.) braziliensis. The few therapeutic drugs available exhibit several limitations, mainly related to drug toxicity and reduced efficacy in some regions. Miltefosine (MF), the only oral drug available for leishmaniasis treatment, is not widely available and has not yet been approved for human use in Brazil. Our group previously reported the existence of differential susceptibility among L. (V.) braziliensis clinical isolates. In this work, we further characterized three of these isolates of L. (V.) braziliensis chosen because they exhibited the lowest and the highest MF half maximal inhibitory concentrations and were therefore considered less tolerant or more tolerant, respectively. Uptake of MF, and also of phosphocholine, were found to be significantly different in more tolerant parasites compared to the less sensitive isolate, which raised the hypothesis of differences in the MF transport complex Miltefosine Transporter (MT)-Ros3. Although some polymorphisms in those genes were found, they did not correlate with the drug susceptibility phenotype. Drug efflux and compartmentalization were similar in the isolates tested, and amphotericin B susceptibility was retained in MF tolerant parasites, suggesting that increased fitness was also not the basis of observed differences. Transcriptomic analysis revealed that Ros3 mRNA levels were upregulated in the sensitive strain compared to the tolerant ones. Increased mRNA abundance in more tolerant isolates was validated by quantitative PCR. Our results suggest that differential gene expression of the MT transporter complex is the basis of the differential susceptibility in these unselected, naturally occurring parasites.


Asunto(s)
Resistencia a Medicamentos , Leishmania braziliensis/efectos de los fármacos , Leishmaniasis Cutánea/parasitología , Fosforilcolina/análogos & derivados , Transporte Biológico , Perfilación de la Expresión Génica , Humanos , Leishmania braziliensis/genética , Pruebas de Sensibilidad Parasitaria , Fosforilcolina/farmacología , Proteínas Protozoarias/genética , Análisis de Secuencia de ARN
6.
Front Microbiol ; 9: 1177, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29922261

RESUMEN

Leishmaniasis is caused by trypanosomatid protozoa of the genus Leishmania, which infect preferentially macrophages. The disease affects 12 million people worldwide, who may present cutaneous, mucocutaneous or visceral forms. Several factors influence the form and severity of the disease, and the main ones are the Leishmania species and the host immune response. CD100 is a membrane bound protein that can also be shed. It was first identified in T lymphocytes and latter shown to be induced in macrophages by inflammatory stimuli. The soluble CD100 (sCD100) reduces migration and expression of inflammatory cytokines in human monocytes and dendritic cells, as well as the intake of oxidized low-density lipoprotein (oxLDL) by human macrophages. Considering the importance of macrophages in Leishmania infection and the potential role of sCD100 in the modulation of macrophage phagocytosis and activation, we analyzed the expression and distribution of CD100 in murine macrophages and the effects of sCD100 on macrophage infection by Leishmania (Leishmania) amazonensis. Here we show that CD100 expression in murine macrophages increases after infection with Leishmania. sCD100 augments infection and phagocytosis of Leishmania (L.) amazonensis promastigotes by macrophages, an effect dependent on macrophage CD72 receptor. Besides, sCD100 enhances phagocytosis of zymosan particles and infection by Trypanosoma cruzi.

7.
Stem Cell Rev Rep ; 14(4): 585-598, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29442223

RESUMEN

The purpose of the present study was to evaluate the neural protein expression pattern of human multipotent mesenchymal stromal cells (hMSCs) treated with forskolin (free-form/FF). The study investigated forskolin's capacity to enhance intracellular levels of cyclic adenosine monophosphate (cAMP) by activating adenylate cyclase and probably by inducing neuron-like cells in vitro. In addition, because nanotechnology is a growing field of tissue engineering, we also assessed the action of a new system called the nanostructured-forskolin (NF) to examine the improvement of drug delivery. Afterwards, the cells were submitted to low-level laser irradiation to evaluate possible photobiostimulatory effects. Investigations using the immunofluorescence by confocal microscopy and Western blot methods revealed the expression of the neuronal marker ß-tubulin III. Fluorescence intensity quantification analysis using INCell Analyzer System for ß-tubulin III was used to examine significant differences. The results showed that after low-level laser irradiation exposure, there was a tendency to increase the ß-tubulin III expression in all groups, as expected in the photobiostimulation process. Notably, this process induced for irradiation was more pronounced in irradiated nanoforskolin cells (INF) compared to non-irradiated free-forskolin control cells (NFFC). However, there was also an increase in ß-tubulin III protein expression in the groups: irradiated nanocontrol cells (INC) compared to non-irradiated free-forskolin control cells (NFF) and after treatment with non-irradiated free-forskolin (NFF) and non-irradiated nanoforskolin (NNFC). We concluded that the methods using low-level laser irradiation and/or nanoparticles showed an up-regulation of neural-protein expression in hMSCs that could be used to facilitate cellular therapy protocols in the near future.


Asunto(s)
Células de la Médula Ósea/efectos de la radiación , Rayos Láser , Células Madre Mesenquimatosas/efectos de la radiación , Neuronas/efectos de la radiación , Tubulina (Proteína)/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/efectos de la radiación , Células Cultivadas , Colforsina/farmacología , Relación Dosis-Respuesta en la Radiación , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Vasodilatadores/farmacología
8.
Exp Cell Res ; 363(2): 271-282, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29355494

RESUMEN

Extracellular matrix (ECM) serves as a reservoir for biologically active factors, such as growth factors and proteases that influence the tumor cell behavior. ADAMTS-1 (a disintegrin and metalloprotease with thrombospondin motifs) is a secreted protease that has the ability to modify the ECM during physiological and pathological processes. Here, we analyzed the role played by ADAMTS-1 regulating HGF and TGF-ß1 activities in the high-grade fibrosarcoma cell line (HT1080). We generated HT1080 and HEK293T cells overexpressing ADAMTS-1. HT1080 cells overexpressing ADAMTS-1 (HT1080-MPA) exhibited a significant decrease in cell proliferation and migration velocity, both in presence of HGF. We obtained similar results with ADAMTS-1-enriched conditioned medium from other cell type. However, ADAMTS-1 overexpression failed to affect TGF-ß1 activity associated with HT1080 cell proliferation and migration velocity. Immunoblotting showed that ADAMTS-1 overexpression disturbs c-Met activation upon HGF stimulation. Downstream ERK1/2 and FAK signaling pathways are also influenced by this protease. Additionally, ADAMTS-1 decreased the size of the fibrosarcospheres, both under normal conditions and in the presence of HGF. Likewise, in presence of HGF, ADAMTS-1 overexpression in HT1080 disrupted microtumors formation in vivo. These microtumors, including individual cells, presented characteristics of non-invasive lesions (rounded morphology). Our results suggest that ADAMTS-1 is involved in regulating HGF-related functions on fibrosarcoma cells. This protease may then represent an endogenous mechanism in controlling the bioavailability of different growth factors that have a direct influence on tumor cell behavior.


Asunto(s)
Proteína ADAMTS1/metabolismo , Proliferación Celular/efectos de los fármacos , Factor de Crecimiento de Hepatocito/farmacología , Proteínas Proto-Oncogénicas c-met/metabolismo , Línea Celular Tumoral , Movimiento Celular/fisiología , Matriz Extracelular/metabolismo , Fibrosarcoma/tratamiento farmacológico , Fibrosarcoma/patología , Células HEK293 , Humanos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo
9.
Front Immunol ; 8: 881, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28824619

RESUMEN

Mesenchymal stromal cells (MSCs) are multipotent cells with abilities to exert immunosuppressive response promoting tissue repair. Studies have shown that MSCs can secrete extracellular vesicles (MVs-MSCs) with similar regulatory functions to the parental cells. Furthermore, strong evidence suggesting that MVs-MSCs can modulate several immune cells (i.e., Th1, Th17, and Foxp3+ T cells). However, their precise effect on macrophages (Mϕs) remains unexplored. We investigated the immunoregulatory effect of MVs-MSCs on activated M1-Mϕs in vitro and in vivo using differentiated bone marrow Mϕs and an acute experimental model of thioglycollate-induced peritonitis, respectively. We observed that MVs-MSCs shared surface molecules with MSCs (CD44, CD105, CD90, CD73) and expressed classical microvesicle markers (Annexin V and CD9). The in vitro treatment with MVs-MSCs exerted a regulatory-like phenotype in M1-Mϕs, which showed higher CD206 level and reduced CCR7 expression. This was associated with decreased levels of inflammatory molecules (IL-1ß, IL-6, nitric oxide) and increased immunoregulatory markers (IL-10 and Arginase) in M1-Mϕs. In addition, we detected that MVs-MSCs promoted the downregulation of inflammatory miRNAs (miR-155 and miR-21), as well as, upregulated its predicted target gene SOCS3 in activated M1-Mϕs. In vivo MVs-MSCs treatment reduced the Mϕs infiltrate in the peritoneal cavity inducing a M2-like regulatory phenotype in peritoneal Mϕs (higher arginase activity and reduced expression of CD86, iNOS, IFN-γ, IL-1ß, TNF-α, IL-1α, and IL-6 molecules). This in vivo immunomodulatory effect of MVs-MSCs on M1-Mϕs was partially associated with the upregulation of CX3CR1 in F4/80+/Ly6C+/CCR2+ Mϕs subsets. In summary, our findings indicate that MVs-MSCs can modulate an internal program in activated Mϕs establishing an alternative regulatory-like phenotype.

10.
Oncotarget ; 7(31): 49998-50016, 2016 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-27374178

RESUMEN

Extracellular vesicles play important roles in tumor development. Many components of these structures, including microvesicles and exosomes, have been defined. However, mechanisms by which extracellular vesicles affect tumor progression are not fully understood. Here, we investigated vesicular communication between mammary carcinoma cells and neighboring nontransformed mammary fibroblasts. Nonbiased proteomic analysis found that over 1% of the entire proteome is represented in these vesicles, with the neuroblast differentiation associated protein AHNAK and annexin A2 being the most abundant. In particular, AHNAK was found to be the most prominent component of these vesicles based on peptide number, and appeared necessary for their formation. In addition, we report here that carcinoma cells produce vesicles that promote the migration of recipient fibroblasts. These data suggest that AHNAK enables mammary carcinoma cells to produce and release extracellular vesicles that cause disruption of the stroma by surrounding fibroblasts. This paradigm reveals fundamental mechanisms by which vesicular communication between carcinoma cells and stromal cells can promote cancer progression in the tumor microenvironment.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinoma/metabolismo , Fibroblastos/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Anexina A2/biosíntesis , Comunicación Celular , Línea Celular Tumoral , Membrana Celular/metabolismo , Movimiento Celular , Proliferación Celular , Cromatografía Liquida , Técnicas de Cocultivo , Exosomas/metabolismo , Humanos , Inmunohistoquímica , Células MCF-7 , Espectrometría de Masas , Microscopía Electrónica de Transmisión , Nanopartículas/química , Proteoma , Proteómica/métodos , Células del Estroma/metabolismo , Microambiente Tumoral
11.
Oncotarget ; 7(30): 47904-47917, 2016 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-27323814

RESUMEN

Laminin peptides influence tumor behavior. In this study, we addressed whether laminin peptide C16 (KAFDITYVRLKF, γ1 chain) would increase invadopodia activity of cells from squamous cell carcinoma (CAL27) and fibrosarcoma (HT1080). We found that C16 stimulates invadopodia activity over time in both cell lines. Rhodamine-conjugated C16 decorates the edge of cells, suggesting a possible binding to membrane receptors. Flow cytometry showed that C16 increases activated ß1 integrin, and ß1 integrin miRNA-mediated depletion diminishes C16-induced invadopodia activity in both cell lines. C16 stimulates Src and ERK 1/2 phosphorylation, and ERK 1/2 inhibition decreases peptide-induced invadopodia activity. C16 also increases cortactin phosphorylation in both cells lines. Based on our findings, we propose that C16 regulates invadopodia activity over time of squamous carcinoma and fibrosarcoma cells, probably through ß1 integrin, Src and ERK 1/2 signaling pathways.


Asunto(s)
Integrina beta1/metabolismo , Laminina/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Podosomas/efectos de los fármacos , Familia-src Quinasas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Laminina/química , Sistema de Señalización de MAP Quinasas/fisiología , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Fragmentos de Péptidos/química , Podosomas/metabolismo , Podosomas/patología , Carcinoma de Células Escamosas de Cabeza y Cuello , Transfección
12.
Sci Rep ; 5: 12425, 2015 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-26213355

RESUMEN

The role of autophagy in cell death is still controversial and a lot of debate has concerned the transition from its pro-survival to its pro-death roles. The similar structure of the triterpenoids Betulinic (BA) and Oleanolic (OA) acids allowed us to prove that this transition involves parallel damage in mitochondria and lysosome. After treating immortalized human skin keratinocytes (HaCaT) with either BA or OA, we evaluated cell viability, proliferation and mechanism of cell death, function and morphology of mitochondria and lysosomes, and the status of the autophagy flux. We also quantified the interactions of BA and OA with membrane mimics, both in-vitro and in-silico. Essentially, OA caused mitochondrial damage that relied on autophagy to rescue cellular homeostasis, which failed upon lysosomal inhibition by Chloroquine or Bafilomycin-A1. BA caused parallel damage on mitochondria and lysosome, turning autophagy into a destructive process. The higher cytotoxicity of BA correlated with its stronger efficiency in damaging membrane mimics. Based on these findings, we underlined the concept that autophagy will turn into a destructive outcome when there is parallel damage in mitochondrial and lysosomal membranes. We trust that this concept will help the development of new drugs against aggressive cancers.


Asunto(s)
Apoptosis/fisiología , Autofagia/fisiología , Queratinocitos/fisiología , Lisosomas/fisiología , Mitocondrias/fisiología , Triterpenos Pentacíclicos/administración & dosificación , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Lisosomas/efectos de los fármacos , Lisosomas/ultraestructura , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura
13.
PLoS One ; 7(12): e51804, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23272170

RESUMEN

Trypanosoma cruzi is a protozoan parasite that comprises different phylogenetic groups and is the causative agent of Chagas' disease. Different T. cruzi strains present differences in infectivity in in vitro and in vivo experimental models, which are likely related to the expression of different virulence factors. Amastin is a surface glycoprotein abundantly expressed on the intracellular mammalian amastigote form of the parasite. In this study, we showed that a highly infective strain (G strain) of extracellular amastigote (EA) invasive forms expressed reduced RNA levels of amastin compared to a less infective strain (CL). The treatment of HeLa cells with recombinant δ-amastin reduced infectivity of EA forms. However, the ectopic expression of δ-amastin accelerated amastigote differentiation into trypomastigotes. Corroborating the virulence behavior in association with amastin expression, the EAs overexpressing amastin were precociously and robustly observed in the liver of susceptible mouse strains (A/JUnib), whereas parasitemia was never detected in in vivo assays. This is the first report on the regulatory role of amastin in the course of both in vitro and in vivo T. cruzi infection.


Asunto(s)
Glicoproteínas de Membrana/genética , Proteínas Protozoarias/genética , Trypanosoma cruzi/genética , Trypanosoma cruzi/patogenicidad , Secuencia de Aminoácidos , Animales , Línea Celular , Enfermedad de Chagas/parasitología , Expresión Génica , Interacciones Huésped-Parásitos , Humanos , Hígado/parasitología , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Ratones , Datos de Secuencia Molecular , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transcripción Genética
14.
PLoS One ; 7(12): e51384, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23251513

RESUMEN

BACKGROUND: P21 is a secreted protein expressed in all developmental stages of Trypanosoma cruzi. The aim of this study was to determine the effect of the recombinant protein based on P21 (P21-His(6)) on inflammatory macrophages during phagocytosis. FINDINGS: Our results showed that P21-His(6) acts as a phagocytosis inducer by binding to CXCR4 chemokine receptor and activating actin polymerization in a way dependent onthe PI3-kinase signaling pathway. CONCLUSIONS: Thus, our results shed light on the notion that native P21 is a component related to T. cruzi evasion from the immune response and that CXCR4 may be involved in phagocytosis. P21-His(6) represents an important experimental control tool to study phagocytosis signaling pathways of different intracellular parasites and particles.


Asunto(s)
Fagocitosis , Proteínas Protozoarias/metabolismo , Trypanosoma cruzi/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Receptores CXCR4/metabolismo , Proteínas Recombinantes/metabolismo
15.
Microbes Infect ; 11(5): 563-70, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19344784

RESUMEN

Trypanosoma cruzi genomic database was screened for hypothetical proteins that showed high probability of being secreted or membrane anchored and thus, likely involved in host-cell invasion. A sequence that codes for a 21kDa protein that showed high probability of being secreted was selected. After cloning this protein sequence, the results showed that it was a ubiquitous protein and secreted by extracellular amastigotes. The recombinant form (P21-His(6)) adhered to HeLa cells in a dose-dependent manner. Pretreatment of host cells with P21-His(6) inhibited cell invasion by extracellular amastigotes from G and CL strains. On the other hand, when the protein was added to host cells at the same time as amastigotes, an increase in cell invasion was observed. Host-cell pretreatment with P21-His(6) augmented invasion by metacyclic trypomastigotes. Moreover, polyclonal antibody anti-P21 inhibited invasion only by extracellular amastigotes and metacyclic trypomastigotes from G strain. These results suggested that P21 might be involved in T. cruzi cell invasion. We hypothesize that P21 could be secreted in the juxtaposition parasite-host cell and triggers signaling events yet unknown that lead to parasite internalization.


Asunto(s)
Proteínas Protozoarias/fisiología , Trypanosoma cruzi/patogenicidad , Factores de Virulencia/fisiología , Animales , Chlorocebus aethiops , ADN Protozoario/química , ADN Protozoario/genética , Células HeLa , Humanos , Datos de Secuencia Molecular , Peso Molecular , Unión Proteica , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Análisis de Secuencia de ADN , Trypanosoma cruzi/genética , Células Vero , Factores de Virulencia/química , Factores de Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...