Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Parasitol Res ; 2024: 1858154, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39131749

RESUMEN

Background: The aim of this study was to assess the anthelmintic activity of Lannea kerstingii and Ficus thonningii, on a nematode model, to promote their use in the Cameroonian pharmacopoeia for the treatment of helminthiases. Methods: One nematode was used, Heligmosomoides polygyrus. First, the effect of the extracts on the eggs and larval stages (L1, L2, and L3) of H. polygyrus was evaluated, 100 µL of extract and 100 µL of parasite suspension (containing 50 eggs) were mixed in a 96-well microplate. The 96-well microplate was incubated for 20 h at 25°C in the WMicroTracker which measures the motility of the worms at various concentrations. Finally, docking studies were conducted by using the Glide module in Schrodinger Maestro. Results: The ethanolic extract of L. kerstingii with the half maximal inhibitory concentration (IC50) of 0.1371 mg/mL produced a higher ovicidal effect than the effect produced by other extracts of these plants. However, with an IC50 of 0.31 mg/mL, the aqueous extract of F. thonningii showed the greatest effect on the L2 stage. The aqueous and ethanolic extracts of L. kerstingii and F. thonningii inhibited the development of the L3 larvae of H. polygyrus with a better effect for the ethanolic extracts. Conclusion: The use of L. kerstingii and F. thonningii for the treatment of helminthiasis has been proved in vitro and in silico by this research. However, more research is required, especially on the acute toxicity and in vivo anthelmintic efficacy to validate this scientific investigation.

2.
J Trop Med ; 2024: 8564163, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38974476

RESUMEN

Background: Helminthiasis is endemic in Chad and constitutes a public health problem, particularly among school-age children. The aim of this study was to evaluate the anthelmintic activity of extracts of Khaya anthotheca and Faidherbia albida used in Chad by traditional healers for the treatment of helminthiasis. Methods: The anthelmintic activity was assessed against Heligmosomoides polygyrus and Caenorhabditis elegans larvae using the Worm Microtracker. Embryonated eggs, L1, L2, and L3 larvae of H. polygyrus were obtained after 24 h, 48 h, and 7 days of coproculture and L4 larvae of C. elegans culture using standard procedures. One hundred microliters of extracts at various concentrations, with albendazole and distilled water were, put in contact with 100 µL of H. polygyrus suspension (containing 50 parasites at various developmental stages) in a microplate and incubated for 20 h at 25°C in the Worm Microtracker. The same procedure was adopted for C. elegans, but with 180 µL of OP50. 19 µL of C. elegans suspension (containing 50 larvae) was put in contact with 1 µL of extract at various concentrations and incubated in the Worm Microtracker. Docking studies were carried out using the Schrodinger Maestro software's Glide module. The score function in the software was used to rank and group distinct possible adduct structures generated by molecular docking. Results: The aqueous and ethanolic extracts of F. albida at a concentration of 2.5 mg/mL showed the same activity as albendazole (100 ± 0.00) on hatching. The IC50s of the aqueous extracts of the two plants (IC50: 0.6212 mg/mL and 0.71 mg/mL, respectively) were comparable on egg hatching of H. polygyrus with no significant difference (p ≥ 0.05) with respect to the ethanol extracts (IC50: 0.70 mg/mL and 0.81 mg/mL, respectively). There was no significant difference between the percentage inhibition of extracts and albendazole on the L1 larvae of H. polygyrus (p ≥ 0.05). The aqueous extracts acted more effectively than the ethanol extracts on the L1 larvae of H. polygyrus with an IC50 of 0.5588 and ∼9.858e - 005 mg/ml, respectively, for K. anthotheca and F. albida. The aqueous extracts of K. anthotheca and F. albida on L3 larvae of H. polygyrus had inhibitory percentages of 92.6 ± 0.62 and 91.37 ± 0.8 at 2.5 mg/mL which were lower than albendazole (100 ± 0.00). The aqueous extracts of K. anthotheca and F. albida on C. elegance showed IC50 of 0.2775 µg/mL and 0.5115 µg/mL, respectively, and were more effective than the ethanol extracts. Examining K. anthotheca and F. albida through the interaction with the protein receptor and its results also confirmed our assumption that the compound used has hydroxyl and carbonyl groups as well as aromatic rings and is exposed to phenolic and flavonoid groups in a more specific way, and it shows a better inhibitory effect. Conclusions: This study scientifically validates the use of extracts of the two plants in the traditional treatment of helminthiasis. However, it will be necessary to evaluate the in vivo anthelmintic activity and toxicity. Examining the ADME properties of these compounds also supports the potential of these ligands to be transformed into pharmaceutical forms.

3.
Immunity ; 57(6): 1260-1273.e7, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38744292

RESUMEN

Upon parasitic helminth infection, activated intestinal tuft cells secrete interleukin-25 (IL-25), which initiates a type 2 immune response during which lamina propria type 2 innate lymphoid cells (ILC2s) produce IL-13. This causes epithelial remodeling, including tuft cell hyperplasia, the function of which is unknown. We identified a cholinergic effector function of tuft cells, which are the only epithelial cells that expressed choline acetyltransferase (ChAT). During parasite infection, mice with epithelial-specific deletion of ChAT had increased worm burden, fitness, and fecal egg counts, even though type 2 immune responses were comparable. Mechanistically, IL-13-amplified tuft cells release acetylcholine (ACh) into the gut lumen. Finally, we demonstrated a direct effect of ACh on worms, which reduced their fecundity via helminth-expressed muscarinic ACh receptors. Thus, tuft cells are sentinels in naive mice, and their amplification upon helminth infection provides an additional type 2 immune response effector function.


Asunto(s)
Acetilcolina , Mucosa Intestinal , Animales , Acetilcolina/metabolismo , Ratones , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/parasitología , Colina O-Acetiltransferasa/metabolismo , Interleucina-13/metabolismo , Interleucina-13/inmunología , Ratones Noqueados , Ratones Endogámicos C57BL , Helmintiasis/inmunología , Helmintiasis/parasitología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Inmunidad Innata , Nematospiroides dubius/inmunología , Células en Penacho
4.
bioRxiv ; 2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-38014296

RESUMEN

The murine helminth parasite Heligmosomoides polygyrus expresses a family of modular proteins which, replicating the functional activity of the immunomodulatory cytokine TGF-ß, have been named TGM (TGF-ß Μimic). Multiple domains bind to different receptors, including TGF-ß receptors TßRI (ALK5) and TßRII through domains 1-3, and prototypic family member TGM1 binds the cell surface co-receptor CD44 through domains 4-5. This allows TGM1 to induce T lymphocyte Foxp3 expression, characteristic of regulatory (Treg) cells, and to activate a range of TGF-ß-responsive cell types. In contrast, a related protein, TGM4, targets a much more restricted cell repertoire, primarily acting on myeloid cells, with less potent effects on T cells and lacking activity on other TGF-ß-responsive cell types. TGM4 binds avidly to myeloid cells by flow cytometry, and can outcompete TGM1 for cell binding. Analysis of receptor binding in comparison to TGM1 reveals a 10-fold higher affinity than TGM1 for TGFßR-I (TßRI), but a 100-fold lower affinity for TßRII through Domain 3. Consequently, TGM4 is more dependent on co-receptor binding; in addition to CD44, TGM4 also engages CD49d (Itga4) through Domains 1-3, as well as CD206 and Neuropilin-1 through Domains 4 and 5. TGM4 was found to effectively modulate macrophage populations, inhibiting lipopolysaccharide-driven inflammatory cytokine production and boosting interleukin (IL)-4-stimulated responses such as Arginase-1 in vitro and in vivo. These results reveal that the modular nature of TGMs has allowed the fine tuning of the binding affinities of the TßR- and co-receptor binding domains to establish cell specificity for TGF-ß signalling in a manner that cannot be attained by the mammalian cytokine.

5.
Proc Natl Acad Sci U S A ; 120(34): e2302370120, 2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37590410

RESUMEN

Long-lived parasites evade host immunity through highly evolved molecular strategies. The murine intestinal helminth, Heligmosomoides polygyrus, down-modulates the host immune system through release of an immunosuppressive TGF-ß mimic, TGM1, which is a divergent member of the CCP (Sushi) protein family. TGM1 comprises 5 domains, of which domains 1-3 (D1/2/3) bind mammalian TGF-ß receptors, acting on T cells to induce Foxp3+ regulatory T cells; however, the roles of domains 4 and 5 (D4/5) remain unknown. We noted that truncated TGM1, lacking D4/5, showed reduced potency. Combination of D1/2/3 and D4/5 as separate proteins did not alter potency, suggesting that a physical linkage is required and that these domains do not deliver an independent signal. Coprecipitation from cells treated with biotinylated D4/5, followed by mass spectrometry, identified the cell surface protein CD44 as a coreceptor for TGM1. Both full-length and D4/5 bound strongly to a range of primary cells and cell lines, to a greater degree than D1/2/3 alone, although some cell lines did not respond to TGM1. Ectopic expression of CD44 in nonresponding cells conferred responsiveness, while genetic depletion of CD44 abolished enhancement by D4/5 and ablated the ability of full-length TGM1 to bind to cell surfaces. Moreover, CD44-deficient T cells showed attenuated induction of Foxp3 by full-length TGM1, to levels similar to those induced by D1/2/3. Hence, a parasite protein known to bind two host cytokine receptor subunits has evolved a third receptor specificity, which serves to raise the avidity and cell type-specific potency of TGF-ß signaling in mammalian cells.


Asunto(s)
Parásitos , Animales , Ratones , Linfocitos T Reguladores , Transducción de Señal , Factor de Crecimiento Transformador beta , Factores de Transcripción Forkhead , Mamíferos
6.
J Virol ; 89(20): 10467-81, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26246581

RESUMEN

UNLABELLED: Bluetongue virus (BTV) is an arbovirus transmitted to livestock by midges of the Culicoides family and is the etiological agent of a hemorrhagic disease in sheep and other ruminants. In mammalian cells, BTV particles are released primarily by virus-induced cell lysis, while in insect cells they bud from the plasma membrane and establish a persistent infection. BTV possesses a ten-segmented double-stranded RNA genome, and NS3 proteins are encoded by segment 10 (Seg-10). The viral nonstructural protein 3 (NS3) plays a key role in mediating BTV egress as well as in impeding the in vitro synthesis of type I interferon in mammalian cells. In this study, we asked whether genetically distant NS3 proteins can alter BTV-host interactions. Using a reverse genetics approach, we showed that, depending on the NS3 considered, BTV replication kinetics varied in mammals but not in insects. In particular, one of the NS3 proteins analyzed harbored a proline at position 24 that leads to its rapid intracellular decay in ovine but not in Culicoides cells and to the attenuation of BTV virulence in a mouse model of disease. Overall, our data reveal that the genetic variability of Seg-10/NS3 differentially modulates BTV replication kinetics in a host-specific manner and highlight the role of the host-specific variation in NS3 protein turnover rate. IMPORTANCE: BTV is the causative agent of a severe disease transmitted between ruminants by biting midges of Culicoides species. NS3, encoded by Seg-10 of the BTV genome, fulfills key roles in BTV infection. As Seg-10 sequences from various BTV strains display genetic variability, we assessed the impact of different Seg-10 and NS3 proteins on BTV infection and host interactions. In this study, we revealed that various Seg-10/NS3 proteins alter BTV replication kinetics in mammals but not in insects. Notably, we found that NS3 protein turnover may vary in ovine but not in Culicoides cells due to a single amino acid residue that, most likely, leads to rapid and host-dependent protein degradation. Overall, this study highlights that genetically distant BTV Seg-10/NS3 influence BTV biological properties in a host-specific manner and increases our understanding of how NS3 proteins contribute to the outcome of BTV infection.


Asunto(s)
Virus de la Lengua Azul/genética , Células Endoteliales/virología , Regulación Viral de la Expresión Génica , Genoma Viral , Proteínas no Estructurales Virales/genética , Replicación Viral/genética , Secuencia de Aminoácidos , Animales , Aorta/metabolismo , Aorta/patología , Aorta/virología , Virus de la Lengua Azul/química , Virus de la Lengua Azul/metabolismo , Línea Celular Transformada , Ceratopogonidae , Plexo Coroideo/metabolismo , Plexo Coroideo/patología , Plexo Coroideo/virología , Cricetulus , Células Endoteliales/metabolismo , Células Endoteliales/patología , Especificidad del Huésped , Ratones , Datos de Secuencia Molecular , Cultivo Primario de Células , Estabilidad Proteica , Proteolisis , Genética Inversa , Ovinos , Transducción de Señal , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Liberación del Virus/genética
7.
PLoS Pathog ; 9(7): e1003440, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23853584

RESUMEN

Influenza A NS1 and NS2 proteins are encoded by the RNA segment 8 of the viral genome. NS1 is a multifunctional protein and a virulence factor while NS2 is involved in nuclear export of viral ribonucleoprotein complexes. A yeast two-hybrid screening strategy was used to identify host factors supporting NS1 and NS2 functions. More than 560 interactions between 79 cellular proteins and NS1 and NS2 proteins from 9 different influenza virus strains have been identified. These interacting proteins are potentially involved in each step of the infectious process and their contribution to viral replication was tested by RNA interference. Validation of the relevance of these host cell proteins for the viral replication cycle revealed that 7 of the 79 NS1 and/or NS2-interacting proteins positively or negatively controlled virus replication. One of the main factors targeted by NS1 of all virus strains was double-stranded RNA binding domain protein family. In particular, adenosine deaminase acting on RNA 1 (ADAR1) appeared as a pro-viral host factor whose expression is necessary for optimal viral protein synthesis and replication. Surprisingly, ADAR1 also appeared as a pro-viral host factor for dengue virus replication and directly interacted with the viral NS3 protein. ADAR1 editing activity was enhanced by both viruses through dengue virus NS3 and influenza virus NS1 proteins, suggesting a similar virus-host co-evolution.


Asunto(s)
Adenosina Desaminasa/metabolismo , Interacciones Huésped-Patógeno , Virus de la Influenza A/fisiología , Proteínas no Estructurales Virales/metabolismo , Factores de Virulencia/metabolismo , Replicación Viral , Adenosina Desaminasa/química , Adenosina Desaminasa/genética , Transporte Biológico , Línea Celular , Virus del Dengue/enzimología , Humanos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/metabolismo , Gripe Humana/patología , Gripe Humana/virología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Recombinantes/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Mucosa Respiratoria/virología , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Especificidad de la Especie , Técnicas del Sistema de Dos Híbridos , Proteínas no Estructurales Virales/genética , Factores de Virulencia/genética
8.
Mol Ther ; 19(9): 1656-66, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21750535

RESUMEN

Identification of new techniques to express proteins into mammal cells is of particular interest for both research and medical purposes. The present study describes the use of engineered vesicles to deliver exogenous proteins into human cells. We show that overexpression of the spike glycoprotein of the vesicular stomatitis virus (VSV-G) in human cells induces the release of fusogenic vesicles named gesicles. Biochemical and functional studies revealed that gesicles incorporated proteins from producer cells and could deliver them to recipient cells. This protein-transduction method allows the direct transport of cytoplasmic, nuclear or surface proteins in target cells. This was demonstrated by showing that the TetR transactivator and the receptor for the murine leukemia virus (MLV) envelope [murine cationic amino acid transporter-1 (mCAT-1)] were efficiently delivered by gesicles in various cell types. We further shows that gesicle-mediated transfer of mCAT-1 confers to human fibroblasts a robust permissiveness to ecotropic vectors, allowing the generation of human-induced pluripotent stem cells in level 2 biosafety facilities. This highlights the great potential of mCAT-1 gesicles to increase the safety of experiments using retro/lentivectors. Besides this, gesicles is a versatile tool highly valuable for the nongenetic delivery of functions such as transcription factors or genome engineering agents.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos , Glicoproteínas de Membrana/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Animales , Canales de Calcio/genética , Canales de Calcio/metabolismo , Biología Computacional , Citometría de Flujo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Inmunohistoquímica , Virus de la Leucemia Murina/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Microscopía Electrónica de Transmisión , Células Madre Pluripotentes/metabolismo , Proteómica/métodos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Factores de Transcripción/genética , Transducción Genética , Vesiculovirus/genética
9.
Physiol Genomics ; 42A(1): 61-70, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20571111

RESUMEN

The adipocyte-derived hormone adiponectin exerts protective actions in several disorders, including some cancers. However, while growing data suggest that adiponectin could be an effective anticancer agent, its mechanism of action in cancer cells is still poorly known. Here, using microarrays, we identified a set of 1,301 genes commonly modulated in three cancer-derived cell lines in response to short-term stimulation with full-length recombinant human adiponectin. Most of these genes are involved in translation regulation, immune or stress responses, and cell proliferation. Furthermore, among genes linked to disease that were retrieved by functional enrichment tests using text mining based on PubMed analysis, we found that 66% are involved in malignant neoplasms, further supporting the link between adiponectin and cancer mechanisms. Bioinformatic analysis demonstrated the diversity of signaling pathways and transcription factors potentially mediating adiponectin effects on gene expression, illustrating the complexity of adiponectin mechanisms of action in cancer cells.


Asunto(s)
Adiponectina/farmacología , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Antineoplásicos/farmacología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/genética , Células HeLa , Células Hep G2 , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Oncol Rep ; 23(2): 523-9, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20043117

RESUMEN

The SNAIL and SLUG transcription factors play important roles in embryogenesis owing to their anti-apoptotic properties and their ability to promote morphogenetic changes by inducing epithelial-mesenchymal transitions (EMT). These characteristics provide many of the proteins in these families with oncogenic and pro-metastatic capabilities when reactivated in cancers. The SCRATCH subgroup of the SNAIL superfamily, including SCRATCH1 and SCRATCH2, display distinct embryonic functions and diverge early in evolution. Despite the described overexpression of SCRT1 (encoding for SCRATCH1) in a small subset of human lung cancers, there is little data supporting a role of SCRATCH proteins in tumorigenesis. To further explore this possibility, we assessed SNAI1 (SNAIL), SNAI2 (SLUG) and SCRT1 (SCRATCH1) expression in a wide panel of human and murine tumors encompassing 151 primary tumors and 6 different cancer types, including melanomas and multiple different carcinomas. Whereas SNAI1 and SNAI2 are widely expressed in human and murine tumors, our results reveal that SCRT1 transcripts are undetectable in nearly all of the examined tumors suggesting that SCRATCH1 plays a minor role, if any, in tumorigenesis. Our data therefore suggest that oncogenic properties are not shared by all SNAIL superfamily members but instead are specifically allotted to the SNAIL subgroup supporting the conclusions that SNAIL and SCRATCH subgroups are functionally divergent and strengthening the hypothesis that the oncogenic potential of SNAIL and SLUG proteins relies on the hijacking of their embryonic functions.


Asunto(s)
Neoplasias/genética , Factores de Transcripción/genética , Animales , Carcinoma/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Mamarias Animales/genética , Melanoma/genética , Ratones , Factores de Transcripción de la Familia Snail , Factores de Transcripción/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...