Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Int J Mol Sci ; 20(23)2019 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-31795474

RESUMEN

Mycobacterium bovis (M. bovis) is the causative agent of bovine tuberculosis in cattle population across the world. Human beings are at equal risk of developing tuberculosis beside a wide range of M. bovis infections in animal species. Autophagic sequestration and degradation of intracellular pathogens is a major innate immune defense mechanism adopted by host cells for the control of intracellular infections. It has been reported previously that the catalytic subunit of protein phosphatase 2A (PP2Ac) is crucial for regulating AMP-activated protein kinase (AMPK)-mediated autophagic signaling pathways, yet its role in tuberculosis is still unclear. Here, we demonstrated that M. bovis infection increased PP2Ac expression in murine macrophages, while nilotinib a tyrosine kinase inhibitor (TKI) significantly suppressed PP2Ac expression. In addition, we observed that TKI-induced AMPK activation was dependent on PP2Ac regulation, indicating the contributory role of PP2Ac towards autophagy induction. Furthermore, we found that the activation of AMPK signaling is vital for the regulating autophagy during M. bovis infection. Finally, the transient inhibition of PP2Ac expression enhanced the inhibitory effect of TKI-nilotinib on intracellular survival and multiplication of M. bovis in macrophages by regulating the host's immune responses. Based on these observations, we suggest that PP2Ac should be exploited as a promising molecular target to intervene in host-pathogen interactions for the development of new therapeutic strategies towards the control of M. bovis infections in humans and animals.


Asunto(s)
Proteínas Quinasas Activadas por AMP/inmunología , Macrófagos/inmunología , Mycobacterium bovis/inmunología , Proteína Fosfatasa 2/inmunología , Tuberculosis/veterinaria , Animales , Autofagia , Bovinos , Interacciones Huésped-Patógeno , Humanos , Macrófagos/microbiología , Ratones , Mycobacterium bovis/fisiología , Fagocitosis , Células RAW 264.7 , Tuberculosis/inmunología , Tuberculosis/microbiología , Tuberculosis Bovina/inmunología , Tuberculosis Bovina/microbiología
2.
Foods ; 9(11)2020 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-33202556

RESUMEN

Healthy diets are necessary for both humans and animals, including poultry. These diets contain various nutrients for maintenance and production in laying hens. Therefore, research was undertaken to explore the efficiency of various dietary flaxseed sources on the n-3 deposition in the egg yolk and gene expression in laying hens. Five dietary groups were analyzed, i.e., (i) a corn-based diet with no flaxseed (FS) as a negative control (NC), (ii) a wheat-based diet supplemented with 10% whole FS without multi-carbohydrase enzymes (MCE) as a positive control (PC), (iii) ground FS supplemented with MCE (FS), (iv) extruded flaxseed meal was supplemented with MCE (EFM), (v) flaxseed oil supplemented with MCE (FSO). Results indicated that egg weight was highest in the NC, FS, EFM, and FSO groups as compared to PC in the 12th week. Egg mass was higher in enzyme supplemented groups as compared to the PC group, but lower than NC. In the 12th week, the HDEP (hen day egg production) was highest in the FS and EFM groups as compared to FSO, PC, and NC. The FCR (feed conversion ratio) was better in enzyme supplemented groups as compared to the PC group. Enzyme addition enhanced the egg quality as compared to PC in the 12th week. The HDL-C (high-density lipoprotein cholesterol) was increased, while LDL-C (low-density lipoprotein cholesterol), VLDL-C (very-low-density lipoprotein cholesterol), TC (total cholesterol), and TG (total triglycerides) were reduced in the enzyme supplemented groups as compared to PC and NC. The FSO deposit more n-3 PUFA and docosahexaenoic acid (DHA) in the egg yolk as compared to FS and EFM groups. The expression of ACOX1, LCPT1, FADS1, FADS2, and ELOV2 genes were upregulated, while PPAR-α was downregulated in the FSO group. The LPL mRNA expression was upregulated in the FS, EFM, and FSO groups as compared to the PC and NC groups. It was inferred that FSO with enzymes at 2.5% is cost-effective, improves the hen performances, upregulated the fatty acid metabolism and ß-oxidation genes expression, and efficiently deposits optimal n-3 PUFA in the egg as per consumer's demand.

3.
Cells ; 8(5)2019 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-31130711

RESUMEN

Nilotinib, a tyrosine kinase inhibitor, has been studied extensively in various tumor models; however, no information exists about the pharmacological action of nilotinib in bacterial infections. Mycobacterium bovis (M. bovis) and Mycobacterium avium subspecies paratuberculosis (MAP) are the etiological agents of bovine tuberculosis and Johne's disease, respectively. Although M. bovis and MAP cause distinct tissue tropism, both of them infect, reside, and replicate in mononuclear phagocytic cells of the infected host. Autophagy is an innate immune defense mechanism for the control of intracellular bacteria, regulated by diverse signaling pathways. Here we demonstrated that nilotinib significantly inhibited the intracellular survival and growth of M. bovis and MAP in macrophages by modulating host immune responses. We showed that nilotinib induced autophagic degradation of intracellular mycobacterium occurred via the inhibition of PI3k/Akt/mTOR axis mediated by abelson (c-ABL) tyrosine kinase. In addition, we observed that nilotinib promoted ubiquitin accumulation around M. bovis through activation of E3 ubiquitin ligase parkin. From in-vivo experiments, we found that nilotinib effectively controlled M. bovis growth and survival through enhanced parkin activity in infected mice. Altogether, our data showed that nilotinib regulates protective innate immune responses against intracellular mycobacterium, both in-vitro and in-vivo, and can be exploited as a novel therapeutic remedy for the control of M. bovis and MAP infections.


Asunto(s)
Autofagia/efectos de los fármacos , Mycobacterium avium subsp. paratuberculosis/efectos de los fármacos , Mycobacterium bovis/efectos de los fármacos , Paratuberculosis/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Tuberculosis Bovina/tratamiento farmacológico , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Bovinos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citoplasma/metabolismo , Citoplasma/microbiología , Femenino , Inmunidad Innata/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteína Oncogénica v-akt/metabolismo , Paratuberculosis/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Proto-Oncogénicas c-abl/metabolismo , Pirimidinas/administración & dosificación , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Tuberculosis Bovina/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA