Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 166
Filtrar
1.
Molecules ; 29(13)2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38999017

RESUMEN

Bimetallic nanostructured catalysts have shown great promise in the areas of energy, environment and magnetics. Tunable composition and electronic configurations due to lattice strain at bimetal interfaces have motivated researchers worldwide to explore them industrial applications. However, to date, the fundamentals of the synthesis of lattice-mismatched bimetallic nanocrystals are still largely uninvestigated for most supported catalyst materials. Therefore, in this work, we have conducted a detailed review of the synthesis and structural characterization of bimetallic nanocatalysts, particularly for renewable energies. In particular, the synthesis of Pt, Au and Pd bimetallic particles in a liquid phase has been critically discussed. The outcome of this review is to provide industrial insights of the rational design of cost-effective nanocatalysts for sustainable conversion technologies.

2.
Can J Physiol Pharmacol ; 101(11): 599-609, 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37459654

RESUMEN

As a common aggressive head and neck cancer, nasopharyngeal carcinoma (NPC) received cisplatin treatment as a first-line chemotherapy. Platinum-induced resistance is a major limitation of current treatment strategy in the advanced NPC. Increased indoleamine 2,3-dioxygenase (IDO1) activities are found in cisplatin-resistant NPC cells versus cisplatin-sensitive NPC cells. As an IDO1 immunosuppressant, NLG-919 has entered clinical phase I to treat advanced solid tumors. To reverse cisplatin resistance, we investigated the combinatory application of cisplatin and NLG-919 in NPC treatment. In vitro biological studies on cisplatin-resistant and cisplatin-sensitive NPC cells were taken to imply that the combination of NLG-919 and cisplatin got a stronger impact on the induction of cell apoptosis and the inhibition of cell migration, exploring superior effect of antitumor over single drug. We proved that the mechanism of the combined therapy could inhibit the activity of IDO1, blocking amino acid tryptophan conversion to kynurenine through the kynurenine pathway, which further inhibited the aryl hydrocarbon receptor expression. Our study underscored the combination of cisplatin and NLG-919 as a potent therapeutic way for the reversal of cisplatin resistance.


Asunto(s)
Cisplatino , Neoplasias Nasofaríngeas , Humanos , Cisplatino/farmacología , Cisplatino/uso terapéutico , Carcinoma Nasofaríngeo/tratamiento farmacológico , Quinurenina/metabolismo , Quinurenina/farmacología , Quinurenina/uso terapéutico , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Hidrocarburo de Aril/uso terapéutico , Transducción de Señal , Neoplasias Nasofaríngeas/tratamiento farmacológico , Neoplasias Nasofaríngeas/patología , Movimiento Celular , Línea Celular Tumoral
3.
FASEB J ; 35(9): e21777, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34403519

RESUMEN

Mycobacterium bovis is the causative agent of bovine tuberculosis and also responsible for serious threat to public health. Koumiss is a fermented mare's milk product, used as traditional drink. Here, we explored the effect of koumiss on gut microbiota and the host immune response against M bovis infection. Therefore, mice were treated with koumiss and fresh mare milk for 14 days before M bovis infection and continue for 5 weeks after infection. The results showed a clear change in the intestinal flora of mice treated with koumiss, and the lungs of mice treated with koumiss showed severe edema, inflammatory infiltration, and pulmonary nodules in M bovis-infected mice. Notably, we found that the content of short-chain fatty acids was significantly lower in the koumiss-treated group compared with the control group. However, the expression of endoplasmic reticulum stress and apoptosis-related proteins in the lungs of koumiss-treated mice were significantly decreased. Collectively, these findings suggest that koumiss treatment disturb the intestinal flora of, which is associated with disease severity and the possible mechanism that induces lungs pathology. Our current findings can be exploited further to establish the "gut-lung" axis which might be a novel strategy for the control of tuberculosis.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Kumis/efectos adversos , Mycobacterium bovis/efectos de los fármacos , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/patología , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Ácidos Grasos/análisis , Heces/química , Heces/microbiología , Femenino , Microbioma Gastrointestinal/inmunología , Caballos , Pulmón/efectos de los fármacos , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Mycobacterium bovis/inmunología , Tuberculosis Pulmonar/dietoterapia , Tuberculosis Pulmonar/metabolismo
4.
J Infect Dis ; 221(3): 438-448, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31495880

RESUMEN

BACKGROUND: Mycobacterium bovis persistently survives in macrophages by developing multiple strategies to evade host immune responses, and the early induction of interferon-ß (IFN-ß) is one of these critical strategies. The mitochondrial transcription factor A (TFAM) plays a vital role in mitochondrial DNA (mtDNA) metabolism and has been suggested to influence IFN-ß production in response to viral infection. However, its role in the production of IFN-ß by M. bovis has not been elucidated. METHODS: In the current study, we investigated the role of TFAM in the production of IFN-ß in M. bovis-infected macrophages. RESULTS: We found that knockdown of TFAM expression significantly reduced M. bovis-induced IFN-ß production, mtDNA copy numbers and cytosolic mtDNA were increased in murine macrophages with M. bovis infection, cytosolic mtDNA contributed to IFN-ß production, and TFAM was required for the increase in mtDNA copy numbers induced by M. bovis. We also observed that TFAM affected the intracellular survival of M. bovis. CONCLUSIONS: Our results suggest that TFAM plays an essential role in M. bovis-induced IFN-ß production by regulating mtDNA copy numbers. This might be a new strategy adopted by M. bovis for its intracellular survival.


Asunto(s)
Replicación del ADN , ADN Mitocondrial/genética , Proteínas de Unión al ADN/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Interferón beta/biosíntesis , Macrófagos/inmunología , Mycobacterium bovis/inmunología , Tuberculosis/inmunología , Tuberculosis/veterinaria , Animales , Línea Celular Tumoral , Citosol/metabolismo , ADN Mitocondrial/metabolismo , Proteínas de Unión al ADN/genética , Técnicas de Silenciamiento del Gen , Proteínas del Grupo de Alta Movilidad/genética , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Mycobacterium bovis/metabolismo , Transducción de Señal/genética , Tuberculosis/microbiología
5.
Neurobiol Dis ; 135: 104704, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31837420

RESUMEN

Evidence of the gut microbiota influencing neurodegenerative diseases has been reported for several neural diseases. However, there is little insight regarding the relationship between the gut microbiota and prion disease. Here, using fecal samples of 12 prion-infected mice and 25 healthy controls, we analyzed the structure of the gut microbiota and metabolic changes by 16S rRNA sequencing and LC-MS-based metabolomics respectively as multi-omic analyses. Additionally, SCFAs and common amino acids were detected by GC-MS and UPLC respectively. Enteric changes induced by prion disease affected both structure and abundances of the gut microbiota. The gut microbiota of infected mice displayed greater numbers of Proteobacteria and less Saccharibacteria at the phylum level and more Lactobacillaceae and Helicobacteraceae and less Prevotellaceae and Ruminococcaceae at the family level. A total of 145 fecal metabolites were found to be significantly different in prion infection, and most (114) of these were lipid metabolites. Using KEGG pathway enrichment analysis, we found that 3 phosphatidylcholine (PC) compounds significantly decreased and 4 hydrophobic bile acids significantly increased. Decreases of 8 types of short-chain acids (SCFAs) and increases of Cys and Tyr and decreases of His, Trp, and Arg were observed in prion infection. Correlation analysis indicated that the gut microbiota changes observed in our study may have been the shared outcome of prion disease. These findings suggest that prion disease can cause significant shifts in the gut microbiota. Certain bacterial taxa can then respond to the resulting change to the enteric environment by causing dramatic shifts in metabolite levels. Our data highlight the health impact of the gut microbiota and related metabolites in prion disease.


Asunto(s)
Bacterias/patogenicidad , Disbiosis/metabolismo , Microbioma Gastrointestinal/fisiología , Enfermedades por Prión/microbiología , Animales , Ácidos y Sales Biliares/análisis , Disbiosis/microbiología , Heces/química , Heces/microbiología , Femenino , Metabolómica/métodos , Ratones Endogámicos C57BL , ARN Ribosómico 16S/genética
6.
J Org Chem ; 85(4): 2504-2511, 2020 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-31910620

RESUMEN

A complementary and general strategy for the oxidative generation of iminyl radicals from the readily available α-imino-oxy acids has been established through silver-catalyzed decarboxylation. To demonstrate its synthesis utility, the direct C-H cyanoalkylation of heterocycles and quinones with cyclic α-imino-oxy acids via the iminyl radical-mediated C-C bond cleavage is developed. This cost-effective method takes place under mild reaction conditions and exhibits a broad substrate scope.

7.
BMC Infect Dis ; 19(1): 1031, 2019 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-31801478

RESUMEN

BACKGROUND: Mycobacterium bovis (M. bovis) is the principal causative agent of bovine tuberculosis; however, it may also cause serious infection in human being. Type I IFN is a key factor in reducing viral multiplication and modulating host immune response against viral infection. However, the regulatory pathways of Type I IFN signaling during M. bovis infection are not yet fully explored. Here, we investigate the role of Type I IFN signaling in the pathogenesis of M. bovis infection in mice. METHODS: C57BL/6 mice were treated with IFNAR1-blocking antibody or Isotype control 24 h before M. bovis infection. After 21 and 84 days of infection, mice were sacrificed and the role of Type I IFN signaling in the pathogenesis of M. bovis was investigated. ELISA and qRT-PCR were performed to detect the expression of Type I IFNs and related genes. Lung lesions induced by M. bovis were assessed by histopathological examination. Viable bacterial count was determined by CFU assay. RESULTS: We observed an abundant expression of Type I IFNs in the serum and lung tissues of M. bovis infected mice. In vivo blockade of Type I IFN signaling reduced the recruitment of neutrophils to the lung tissue, mediated the activation of macrophages leading to an increased pro-inflammatory profile and regulated the inflammatory cytokine production. However, no impact was observed on T cell activation and recruitment in the early acute phase of infection. Additionally, blocking of type I IFN signaling reduced bacterial burden in the infected mice as compared to untreated infected mice. CONCLUSIONS: Altogether, our results reveal that Type I IFN mediates a balance between M. bovis-mediated inflammatory reaction and host defense mechanism. Thus, modulating Type I IFN signaling could be exploited as a therapeutic strategy against a large repertoire of inflammatory disorders including tuberculosis.


Asunto(s)
Interferón Tipo I/metabolismo , Mycobacterium bovis/patogenicidad , Tuberculosis/tratamiento farmacológico , Tuberculosis/metabolismo , Animales , Anticuerpos/farmacología , Citocinas/metabolismo , Femenino , Humanos , Interferón Tipo I/antagonistas & inhibidores , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Ratones Endogámicos C57BL , Mycobacterium bovis/inmunología , Receptor de Interferón alfa y beta/antagonistas & inhibidores , Receptor de Interferón alfa y beta/inmunología , Transducción de Señal/efectos de los fármacos
8.
Appl Opt ; 58(10): 2463-2470, 2019 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-31045038

RESUMEN

We theoretically investigate the optical bistability in a composite photonic molecule cavity optomechanical system consisting of two whispering gallery mode microcavities, where one of the optical cavities is optomechanical with a high quality factor, and the other optical cavity is an auxiliary cavity with high cavity dissipation. By controlling the coupling strength J between the two cavities determined by their distance, the decay rate ratio δ of the two cavities, and the pump power P, the optical bistability can be controlled. Further, the transmission spectrum of the signal field can be efficiently attenuated or amplified, depending on the power of a second "gating" (pump) field P, and other parameters. Our study for photonic-molecule optomechanics systems may be a promising candidate for single-photon transistors and pave the way for potential applications in quantum information technologies.

9.
Int J Mol Sci ; 20(4)2019 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-30791397

RESUMEN

Cyclic GMP-AMP synthase (cGAS) is an important cytosolic DNA sensor that plays a crucial role in triggering STING-dependent signal and inducing type I interferons (IFNs). cGAS is important for intracellular bacterial recognition and innate immune responses. However, the regulating effect of the cGAS pathway for bone marrow-derived dendritic cells (BMDCs) during Mycobacterium bovis (M. bovis) infection is still unknown. We hypothesized that the maturation and activation of BMDCs were modulated by the cGAS/STING/TBK1/IRF3 signaling pathway. In this study, we found that M. bovis promoted phenotypic maturation and functional activation of BMDCs via the cGAS signaling pathway, with the type I IFN and its receptor (IFNAR) contributing. Additionally, we showed that the type I IFN pathway promoted CD4⁺ T cells' proliferation with BMDC during M. bovis infection. Meanwhile, the related cytokines increased the expression involved in this signaling pathway. These data highlight the mechanism of the cGAS and type I IFN pathway in regulating the maturation and activation of BMDCs, emphasizing the important role of this signaling pathway and BMDCs against M. bovis. This study provides new insight into the interaction between cGAS and dendritic cells (DCs), which could be considered in the development of new drugs and vaccines against tuberculosis.


Asunto(s)
Células Dendríticas/inmunología , Factor 3 Regulador del Interferón/metabolismo , Proteínas de la Membrana/metabolismo , Mycobacterium bovis , Nucleotidiltransferasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Tuberculosis Bovina/inmunología , Tuberculosis Bovina/metabolismo , Animales , Bovinos , Diferenciación Celular , Células Dendríticas/metabolismo , Femenino , Interacciones Huésped-Patógeno , Interferón Tipo I/metabolismo , Ratones , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo , Tuberculosis Bovina/microbiología
10.
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
11.
Int J Mol Sci ; 20(5)2019 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-30845718

RESUMEN

Transcription factors play a significant role during the symptomatic onset and progression of prion diseases. We previously showed the immunomodulatory and nuclear factor of activated T cells' (NFAT) suppressive effects of an immunosuppressant, FK506, in the symptomatic stage and an antibiotic, minocycline, in the pre-symptomatic stage of prion infection in hamsters. Here we used for the first time, a combinatory FK506+minocycline treatment to test its transcriptional modulating effects in the symptomatic stage of prion infection. Our results indicate that prolonged treatment with FK506+minocycline was effective in alleviating astrogliosis and neuronal death triggered by misfolded prions. Specifically, the combinatory therapy with FK506+minocycline lowered the expression of the astrocytes activation marker GFAP and of the microglial activation marker IBA-1, subsequently reducing the level of pro-inflammatory cytokines interleukin 1 beta (IL-1ß) and tumor necrosis factor alpha (TNF-α), and increasing the levels of anti-inflammatory cytokines IL-10 and IL-27. We further found that FK506+minocycline treatment inhibited mitogen-activated protein kinase (MAPK) p38 phosphorylation, NF-kB nuclear translocation, caspase expression, and enhanced phosphorylated cAMP response element-binding protein (pCREB) and phosphorylated Bcl2-associated death promoter (pBAD) levels to reduce cognitive impairment and apoptosis. Interestingly, FK506+minocycline reduced mitochondrial fragmentation and promoted nuclear factor⁻erythroid2-related factor-2 (NRF2)-heme oxygenase 1 (HO-1) pathway to enhance survival. Taken together, our results show that a therapeutic cocktail of FK506+minocycline is an attractive candidate for prolonged use in prion diseases and we encourage its further clinical development as a possible treatment for this disease.


Asunto(s)
Minociclina/administración & dosificación , Enfermedades por Prión/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Tacrolimus/administración & dosificación , Animales , Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , Cricetinae , Modelos Animales de Enfermedad , Regulación hacia Abajo , Quimioterapia Combinada , Proteína Ácida Fibrilar de la Glía/metabolismo , Minociclina/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Enfermedades por Prión/inmunología , Enfermedades por Prión/metabolismo , Tacrolimus/farmacología
12.
Cell Mol Life Sci ; 74(6): 1061-1074, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27682820

RESUMEN

Accumulation of misfolded/unfolded aggregated proteins in the brain is a hallmark of many neurodegenerative diseases affecting humans and animals. Dysregulation of calcium (Ca2+) and disruption of fast axonal transport (FAT) are early pathological events that lead to loss of synaptic integrity and axonal degeneration in early stages of neurodegenerative diseases. Dysregulated Ca2+ in the brain is triggered by accumulation of misfolded/unfolded aggregated proteins in the endoplasmic reticulum (ER), a major Ca2+ storing organelle, ultimately leading to neuronal dysfunction and apoptosis. Calcineurin (CaN), a Ca2+/calmodulin-dependent serine/threonine phosphatase, has been implicated in T cells activation through the induction of nuclear factor of activated T cells (NFAT). In addition to the involvement of several other signaling cascades, CaN has been shown to play a role in early synaptic dysfunction and neuronal death. Therefore, inhibiting hyperactivated CaN in early stages of disease might be a promising therapeutic strategy for treating patients with protein misfolding diseases. In this review, we briefly summarize the structure of CaN, inhibition mechanisms by which immunosuppressants inhibit CaN, role of CaN in maintaining neuronal and synaptic integrity and homeostasis and the role played by CaN in protein unfolding/misfolding neurodegenerative diseases.


Asunto(s)
Calcineurina/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Deficiencias en la Proteostasis/metabolismo , Animales , Calcineurina/química , Calcio/metabolismo , Humanos , Inmunosupresores/metabolismo , Transducción de Señal
13.
Int J Mol Sci ; 20(1)2018 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-30577452

RESUMEN

It is widely accepted that different strains of Mycobacterium tuberculosis have variable degrees of pathogenicity and induce different immune responses in infected hosts. Similarly, different strains of Mycobacterium bovis have been identified but there is a lack of information regarding the degree of pathogenicity of these strains and their ability to provoke host immune responses. Therefore, in the current study, we used a mouse model to evaluate various factors involved in the severity of disease progression and the induction of immune responses by two strains of M. bovis isolated from cattle. Mice were infected with both strains of M. bovis at different colony-forming unit (CFU) via inhalation. Gross and histological findings revealed more severe lesions in the lung and spleen of mice infected with M. bovis N strain than those infected with M. bovis C68004 strain. In addition, high levels of interferon-γ (IFN-γ), interleukin-17 (IL-17), and IL-22 production were observed in the serum samples of mice infected with M. bovis N strain. Comparative genomic analysis showed the existence of 750 single nucleotide polymorphisms and 145 small insertions/deletions between the two strains. After matching with the Virulence Factors Database, mutations were found in 29 genes, which relate to 17 virulence factors. Moreover, we found an increased number of virulent factors in M. bovis N strain as compared to M. bovis C68004 strain. Taken together, our data reveal that variation in the level of pathogenicity is due to the mutation in the virulence factors of M. bovis N strain. Therefore, a better understanding of the mechanisms of mutation in the virulence factors will ultimately contribute to the development of new strategies for the control of M. bovis infection.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Mycobacterium bovis , Tuberculosis Bovina/genética , Tuberculosis Bovina/microbiología , Animales , Biopsia , Bovinos , Citocinas/sangre , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Patógeno/genética , Pulmón/patología , Ratones , Tipificación de Secuencias Multilocus , Mutación , Mycobacterium bovis/clasificación , Mycobacterium bovis/genética , Mycobacterium bovis/inmunología , Mycobacterium bovis/patogenicidad , Polimorfismo de Nucleótido Simple , Bazo/patología , Tuberculosis Bovina/inmunología , Virulencia/genética , Factores de Virulencia
14.
Cell Mol Neurobiol ; 37(4): 717-728, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27430567

RESUMEN

Transmissible spongiform encephalopathies (TSEs) are caused by the accumulation of the abnormal prion protein scrapie (PrPSc). Prion protein aggregation, misfolding, and cytotoxicity in the brain are the major causes of neuronal dysfunction and ultimate neurodegeneration in all TSEs. Parkin, an E3 ubiquitin ligase, has been studied extensively in all major protein misfolding aggregating diseases, especially Parkinson's disease and Alzheimer's disease, but the role of parkin in TSEs remains unknown. Here we investigated the role of parkin in a prion disease cell model in which neuroblastoma2a (N2a) cells were treated with prion peptide PrP106-126. We observed a gradual decrease in the soluble parkin level upon treatment with PrP106-126 in a time-dependent manner. Furthermore, endogenous parkin colocalized with FITC-tagged prion fragment106-126. Overexpression of parkin in N2a cells via transfection repressed apoptosis by enhancing autophagy. Parkin-overexpressing cells also showed reductions in apoptotic BAX translocation to the mitochondria and cytochrome c release to the cytosol, which ultimately inhibited activation of proapoptotic caspases. Taken together, our findings reveal a parkin-mediated cytoprotective mechanism against PrP106-126 toxicity, which is a novel potential therapeutic target for treating prion diseases.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia , Mitocondrias/efectos de los fármacos , Neuroblastoma/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Autofagia/efectos de los fármacos , Autofagia/fisiología , Caspasas/efectos de los fármacos , Caspasas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Citocromos c/metabolismo , Ratones , Mitocondrias/metabolismo , Neuroblastoma/tratamiento farmacológico , Fragmentos de Péptidos/farmacología , Priones/metabolismo , Proteínas Recombinantes/farmacología , Ubiquitina-Proteína Ligasas/genética
15.
Dis Aquat Organ ; 122(3): 195-203, 2017 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-28117298

RESUMEN

A captive 8 yr old male bottlenose dolphin Tursiops truncatus succumbed to septicemia with multisystemic inflammation including suppurative enteritis, encephalitis, and pneumonia with chronic pancreatitis. A pure culture of beta-hemolytic, catalase- and oxidase-negative, Gram-positive cocci was isolated from the hilar lymph nodes and pancreas. The isolate was identified by 16S rDNA sequencing as Streptococcus iniae. Histological examination of the digestive system revealed a mixed infection of both bacteria and fungus. Recognized as a pathogen in fish, dolphins, and humans, this is the first report of S. iniae in a dolphin in mainland China. As the number of managed animals in oceanariums is increasing, so is the frequency of contact with fish used as food for marine mammals and humans, highlighting the importance of education and appropriate personal protective protocols to minimize the risk of transmission. An understanding of marine mammal infectious disease organisms is essential to ensuring the health of marine mammals and humans coming into contact with such animals and their food. This study illustrates a systematic clinical, microbiological, and pathological investigation into a septicemic bottlenose dolphin infected with S. iniae. Our findings provide useful information for those involved in the diagnosis and control of infectious diseases in marine mammals and offer insight into an important zoonotic pathogen.


Asunto(s)
Delfín Mular , Sepsis/veterinaria , Infecciones Estreptocócicas/veterinaria , Streptococcus iniae/aislamiento & purificación , Animales , Resultado Fatal , Masculino , Sepsis/microbiología , Infecciones Estreptocócicas/microbiología
16.
Int J Mol Sci ; 18(12)2017 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-29258190

RESUMEN

Tuberculosis (TB) is a major health threat to the human population worldwide. The etiology of the disease is Mycobacterium tuberculosis (Mtb), a highly successful intracellular pathogen. It has the ability to manipulate the host immune response and to make the intracellular environment suitable for its survival. Many studies have addressed the interactions between the bacteria and the host immune cells as involving many immune mediators and other cellular players. Interferon-ß (IFN-ß) signaling is crucial for inducing the host innate immune response and it is an important determinant in the fate of mycobacterial infection. The role of IFN-ß in protection against viral infections is well established and has been studied for decades, but its role in mycobacterial infections remains much more complicated and debatable. The involvement of IFN-ß in immune evasion mechanisms adopted by Mtb has been an important area of investigation in recent years. These advances have widened our understanding of the pro-bacterial role of IFN-ß in host-pathogen interactions. This pro-bacterial activity of IFN-ß appears to be correlated with its anti-inflammatory characteristics, primarily by antagonizing the production and function of interleukin 1ß (IL-1ß) and interleukin 18 (IL-18) through increased interleukin 10 (IL-10) production and by inhibiting the nucleotide-binding domain and leucine-rich repeat protein-3 (NLRP3) inflammasome. Furthermore, it also fails to provoke a proper T helper 1 (Th1) response and reduces the expression of major histocompatibility complex II (MHC-II) and interferon-γ receptors (IFNGRs). Here we will review some studies to provide a paradigm for the induction, regulation, and role of IFN-ß in mycobacterial infection. Indeed, recent studies suggest that IFN-ß plays a role in Mtb survival in host cells and its downregulation may be a useful therapeutic strategy to control Mtb infection.


Asunto(s)
Interferón beta/metabolismo , Tuberculosis/metabolismo , Animales , Interacciones Huésped-Patógeno , Humanos , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
17.
Cell Commun Signal ; 14(1): 29, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27905994

RESUMEN

Mycobacterium avium subsp. paratuberculosis (MAP) is an intracellular pathogen and is the causative agent of Johne's disease of domestic and wild ruminants. Johne's disease is characterized by chronic granulomatous enteritis leading to substantial economic losses to the livestock sector across the world. MAP persistently survives in phagocytic cells, most commonly in macrophages by disrupting its early antibacterial activity. MAP triggers several signaling pathways after attachment to pathogen recognition receptors (PRRs) of phagocytic cells. MAP adopts a survival strategy to escape the host defence mechanisms via the activation of mitogen-activated protein kinase (MAPK) pathway. The signaling mechanism initiated through toll like receptor 2 (TLR2) activates MAPK-p38 results in up-regulation of interleukin-10 (IL-10), and subsequent repression of inflammatory cytokines. The anti-inflammatory response of IL-10 is mediated through membrane-bound IL-10 receptors, leading to trans-phosphorylation and activation of Janus Kinase (JAK) family receptor-associated tyrosine kinases (TyKs), that promotes the activation of latent transcription factors, signal transducer and activators of transcription 3 (STAT3). IL-10 is an important inhibitory cytokine playing its role in blocking phagosome maturation and apoptosis. In the current review, we describe the importance of IL-10 in early phases of the MAP infection and regulatory mechanisms of the IL-10 dependent pathways in paratuberculosis. We also highlight the strategies to target IL-10, MAPK and STAT3 in other infections caused by intracellular pathogens.


Asunto(s)
Interleucina-10/inmunología , Mycobacterium avium subsp. paratuberculosis/inmunología , Paratuberculosis/inmunología , Animales , Interleucina-10/genética , Quinasas Janus/inmunología , MicroARNs/genética , Proteínas Quinasas Activadas por Mitógenos/inmunología , FN-kappa B/inmunología , Paratuberculosis/genética , Rumiantes , Factores de Transcripción STAT/inmunología , Transducción de Señal , Receptor Toll-Like 2/inmunología
18.
J Neurochem ; 133(5): 722-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25810062

RESUMEN

The formation of neurotoxic prion protein (PrP) oligomers is thought to be a key step in the development of prion diseases. Recently, it was determined that the sonication and shaking of recombinant PrP can convert PrP monomers into ß-state oligomers. Herein, we demonstrate that ß-state oligomeric PrP can be generated through protein misfolding cyclic amplification from recombinant full-length hamster, human, rabbit, and mutated rabbit PrP, and that these oligomers can be used for subsequent research into the mechanisms of PrP-induced neurotoxicity. We have characterized protein misfolding cyclic amplification-induced monomer-to-oligomer conversion of PrP from three species using western blotting, circular dichroism, size-exclusion chromatography, and resistance to proteinase K (PK) digestion. We have further shown that all of the resulting ß-oligomers are toxic to primary mouse cortical neurons independent of the presence of PrP(C) in the neurons, whereas the corresponding monomeric PrP were not toxic. In addition, we found that this toxicity is the result of oligomer-induced apoptosis via regulation of Bcl-2, Bax, and caspase-3 in both wild-type and PrP(-/-) cortical neurons. It is our hope that these results may contribute to our understanding of prion transformation within the brain. We found that ß-state oligomeric PrPs can be generated through protein misfolding cyclic amplification (PMCA) from recombinant full-length hamster, human, rabbit, and mutated rabbit PrP. ß-oligomers are toxic to primary mouse cortical neurons independent of the presence of PrP(C) in the neurons, while the corresponding monomeric PrPs were not toxic. This toxicity is the result of oligomers-induced apoptosis via regulation of Bcl-2, Bax, and caspase-3. These results may contribute to our understanding of prion transformation within the brain.


Asunto(s)
Apoptosis/efectos de los fármacos , Neuronas/efectos de los fármacos , Priones/metabolismo , Priones/farmacología , Deficiencias en la Proteostasis/genética , Proteínas Recombinantes/farmacología , Animales , Caspasa 3/metabolismo , Cricetinae , Endopeptidasa K/química , Amplificación de Genes , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/genética , Conejos , Proteínas Recombinantes/metabolismo , Proteína X Asociada a bcl-2/biosíntesis , Proteína X Asociada a bcl-2/genética
19.
Antonie Van Leeuwenhoek ; 108(1): 163-71, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25980833

RESUMEN

Mycobacterium bovis is the causative agent of tuberculosis in cattle. Infection of macrophages with M. bovis leads to the activation of the "nucleotide binding and oligomerization, leucine-rich repeat and pyrin domains-containing protein 3" (NLRP3) and "absent in melanoma 2" (AIM2) inflammasomes, which in turn triggers release of the proinflammatory cytokine interleukin-1ß (IL-1ß) that contributes to bacterial clearance and plays a crucial role in the host defense. However, NLRP3 and AIM2 inflammasome activation is influenced by several factors and how IL-1ß secretion by M. bovis-infected macrophages is regulated via the inflammasome pathway remains unclear. Here we found that IL-1ß secretion and pro-IL-1ß protein accumulation were inhibited in THP-1 macrophages upon exposure to the virulent M. bovis Beijing strain in the presence of high K(+) concentrations, cycloheximide (a protein synthesis inhibitor) and PR-619 (a deubiquitinating enzyme inhibitor). Scavenging reactive oxygen species (ROS) induced by N-acetylcysteine reduced IL-1ß release independent of the mitochondrial permeability transition. Collectively, our results suggest that IL-1ß secretion by M. bovis-infected THP-1 macrophages is reduced by high extracellular K(+) concentration, inhibition of new protein synthesis, deubiquitination, and ROS generation.


Asunto(s)
Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/inmunología , Macrófagos/microbiología , Mycobacterium bovis/inmunología , Animales , Bovinos , Línea Celular , Cicloheximida/metabolismo , Humanos , Potasio/metabolismo , Especies Reactivas de Oxígeno/metabolismo
20.
Neurodegener Dis ; 15(4): 193-201, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26022183

RESUMEN

Prion diseases are known as neurodegenerative diseases of the central nervous system with a long incubation period. Alzheimer's disease (AD) and prion diseases share the hallmark of severe neuronal loss, although their pathogenic mechanisms are similarly incomplete. It appears that these two neurodegenerative diseases share a complex deterioration of function involved in the onset of neuronal loss. To investigate presymptomatic biochemical changes indicative of the initial stage of prion diseases and decipher the pathophysiological mechanisms of these two neurodegenerative diseases, we performed a differential proteomic analysis on brain tissues of 263K-infected hamsters during the presymptomatic period and transgenic APPSWE, PSEN1dE9 mice (a mouse model of AD). We identified 7 differentially expressed proteins including the ß-soluble N-ethylmaleimide-sensitive factor attachment protein (ß-SNAP) by 2-dimensional gel electrophoresis and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. The ß-SNAP expression patterns in the brains of cases and controls were further quantified by Western blotting. ß-SNAP showed an early decrease followed by a progressive depletion. The expression of ß-SNAP was also significantly downregulated in the mouse model of AD. ß-SNAP is brain-specific and known to bind to the SNAP receptors and is therefore involved in the control of neurotransmitter release as well as in constitutive vesicular transport. Our results suggest that presynaptic failure and abnormalities in neurotransmission may be early events in the development of neuronal dysfunction.


Asunto(s)
Encéfalo/metabolismo , Enfermedades por Prión/metabolismo , Proteínas Solubles de Unión al Factor Sensible a la N-Etilmaleimida/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Cricetinae , Regulación hacia Abajo , Femenino , Ratones , Ratones Transgénicos , Presenilina-1/genética , Proteómica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA