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

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Avian Pathol ; 53(1): 68-79, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37855868

RESUMEN

RESEARCH HIGHLIGHTS: MG-HS regulates the expression of transcription factor STAT5.Transcription factor STAT5 can target miR-33-5p promoter element.MG-influenced STAT5 regulates miR-33-5p and its target gene expression.


Asunto(s)
MicroARNs , Infecciones por Mycoplasma , Mycoplasma gallisepticum , Animales , Mycoplasma gallisepticum/genética , MicroARNs/genética , MicroARNs/metabolismo , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Línea Celular , Infecciones por Mycoplasma/veterinaria , Fibroblastos , Pollos/genética
2.
J Infect Dis ; 2023 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-37929888

RESUMEN

Extracellular vesicles (EVs) mediate intercellular communication by transporting proteins. To investigate the pathogenesis of Mycoplasma gallisepticum (MG), a major threat to the poultry industry, we isolated and characterized MG-produced EVs. Our study highlights the significant impact of MG-derived EVs on immune function and macrophage apoptosis, setting them apart from other MG metabolites. These EVs dose-dependently enhance MG adhesion and proliferation, simultaneously modulating TLR2 and IFN-γ pathways, thereby inhibiting macrophage activation. A comprehensive protein analysis revealed 117 proteins in MG-derived EVs, including established virulence factors such as GapA, CrmA, VlhA, and CrmB. Crucially, these EV-associated proteins significantly contribute to MG infection. Our findings advance our comprehension of MG pathogenesis, offering insights for preventive strategies, and emphasize the pivotal role of MG-derived EVs and their associated proteins. This research sheds light on the composition and crucial role of MG-derived EVs in MG pathogenesis, aiding our fight against MG infections.

3.
Vet Res ; 54(1): 50, 2023 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-37337278

RESUMEN

Chick embryos are a valuable model for studying immunity and vaccines. Therefore, it is crucial to investigate the molecular mechanism of the Mycoplasma gallisepticum (MG)-induced immune response in chick embryos for the prevention and control of MG. In this study, we screened for downregulated let-7d microRNA in MG-infected chicken embryonic lungs to explore its involvement in the innate immune mechanism against MG. Here, we demonstrated that low levels of let-7d are a protective mechanism for chicken embryo primary type II pneumocytes (CP-II) in the presence of MG. Specifically, we found that depressed levels of let-7 in CP-II cells reduced the adhesion capacity of MG. This suppressive effect was achieved through the activated mitogen-activated protein kinase phosphatase 1 (MKP1) target gene and the inactivated mitogen-activated protein kinase (MAPK) pathway. Furthermore, MG-induced hyperinflammation and cell death were both alleviated by downregulation of let-7d. In conclusion, chick embryos protect themselves against MG infection through the innate immune molecule let-7d, which may result from its function as an inhibitor of the MAPK pathway to effectively mitigate MG adhesion, the inflammatory response and cell apoptosis. This study may provide new insight into the development of vaccines against MG.


Asunto(s)
MicroARNs , Mycoplasma gallisepticum , Embrión de Pollo , Animales , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Quinasas Activadas por Mitógenos , Pollos/genética , Inmunidad Innata
4.
Vet Res ; 53(1): 103, 2022 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-36471418

RESUMEN

A disruption in the expression of gga-miR-365-3p was confirmed in the Mycoplasma gallisepticum (MG)-infected Chicken primary alveolar type II epithelial (CP-II) cells based on previous sequencing results, but the role it plays in the infection was unclear. In the present study, we demonstrate that MG evaded cellular host immunity via a gga-miR-365-3p/SOCS5-JAK/STATs negative feedback loop. Specifically, we found that at the initial stage of MG infection in cells, gga-miR-365-3p was rapidly increased and activated the JAK/STAT signaling pathway by inhibiting SOCS5, which induced the secretion of inflammatory factors and triggered immune response against MG infection. Over time, though, the infection progressed, MG gradually destroyed the immune defences of CP-II cells. In late stages of infection, MG escaped host immunity by reducing intracellular gga-miR-365-3p and inhibiting the JAK/STAT pathway to suppress the secretion of inflammatory factors and promote MG adhesion or invasion. These results revealed the game between MG and host cell interactions, providing a new perspective to gain insight into the pathogenic mechanisms of MG or other pathogens. Meanwhile, they also contributed to novel thoughts on the prevention and control of MG and other pathogenic infections, shedding light on the immune modulating response triggered by pathogen invasion and their molecular targeting.


Asunto(s)
MicroARNs , Mycoplasma gallisepticum , Animales , Mycoplasma gallisepticum/genética , Quinasas Janus/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Fibroblastos/metabolismo , Transducción de Señal , Factores de Transcripción STAT/metabolismo , Inmunidad
5.
Microb Pathog ; 155: 104927, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33932542

RESUMEN

Mycoplasma gallisepticum (MG) is a major poultry pathogen that can induce Chronic Respiratory Disease (CRD) in chickens, causing serious economic losses in the poultry industry worldwide. Increasing evidence suggests that microRNAs (miRNAs) act as a vital role in resisting microbial pathogenesis and maintaining cellular mechanism. Our previous miRNAs sequencing data showed that gga-miR-223 expression level significantly decreased in MG-infected chicken lungs. The aim of this study was to reveal the role of gga-miR-223 in MG-induced CRD progression. We found that gga-miR-223 was remarkably down regulated and forkhead box O3 (FOXO3) was up-regulated in both MG-infected chicken embryos lungs and the chicken embryonic fibroblast cell line (DF-1) by qPCR. FOXO3 was verified as the target gene of gga-miR-223 through bioinformatics analysis and dual-luciferase reporter assay. Further studies showed that overexpressed gga-miR-223 could promote cell proliferation, cell cycle, and inhibit cell apoptosis by notably promoting the expression of cell cycle marker genes cyclin-dependent kinase 1 (CDK1), cyclin-dependent kinase 6 (CDK6) and Cyclin D1 (CCND1) and inhibiting the expression of apoptosis markers Bcl-2-like 11(BIM), FAS ligand (FASLG) and TNF-related apoptosis-inducing ligand (TRAIL). As expected, FOXO3 knockdown group got similar results. Overexpression of gga-miR-223 observably promoted cell multiplication, cell cycle progression, and inhibited apoptosis of MG-infected DF-1 cells, while inhibited gga-miR-223 had the opposite effect. Taken together, upon MG-infection, downregulated gga-miR-223 could decrease proliferation, cycle progression, and increase apoptosis through directly targeting FOXO3 to exert an aggravating MG-infectious effect.


Asunto(s)
MicroARNs , Mycoplasma gallisepticum , Animales , Apoptosis , Proliferación Celular , Embrión de Pollo , Pollos , Fibroblastos , MicroARNs/genética
6.
Inflamm Res ; 70(10-12): 1217-1231, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34554275

RESUMEN

OBJECTIVE: Mycoplasma gallisepticum (MG), a notorious avian pathogen, leads to considerable economic losses in the poultry industry. MG infection is characterized by severe, uncontrollable inflammation and host DNA damage. Micro ribonucleic acids (miRNAs) have emerged as important regulators in microbial pathogenesis. However, the role of miRNAs in MG infection is poorly characterized. In this study, we validated the functional roles of gga-miR-142-3p. METHODS: The relative expression of gga-miR-142-3p in the lungs of the MG-infected chicken embryos and the MG-infected chicken embryonic fibroblast cell line (DF-1) was determined by reverse transcription quantitative real-time PCR analysis. Bioinformatics database was used to analysis the target gene of gga-miR-142-3p. The luciferase reporter assay as well as gene expression analysis were conducted to validate the target gene. To further explore the biological functions of gga-miR-142-3p upon MG infection, the cell proliferation was quantified using Cell Counting Kit-8 (CCK-8). Meanwhile, cell cycle analysis and apoptosis were measured using a flow cytometer. RESULTS: gga-miR-142-3p was significantly upregulated in both MG-infected chicken-embryo lungs and the DF-1 cells. gga-miR-142-3p over expression significantly downregulated the expression of pro-inflammatory cytokines, including interleukin-1ß, interleukin-6 and tumor necrosis factor alpha after MG infection. Meanwhile, gga-miR-142-3p enhanced the host defense against MG infection by facilitating cell proliferation, promoting cell progression and inhibiting cell apoptosis. Interestingly, TAB2 knockdown groups show similar results, whereas, TAB2 over-expression groups and gga-miR-142-3p inhibitor groups had thoroughly opposite results. The expression of p-p65 in nuclear factor kappa B (NF-κB) and p-p38 in the mitogen-activated protein kinase (MAPK) pathway was decreased when gga-miR-142-3p was over-expressed. CONCLUSION: Upon MG infection, upregulation of gga-miR-142-3p alleviates inflammation by negatively regulating the signaling pathways of NF-κB and MAPKs by targeting TAB2 and facilitates cell proliferation by inhibiting cell apoptosis and promoting cell cycle progression to defend against MG infection.


Asunto(s)
MicroARNs , Infecciones por Mycoplasma/genética , Infecciones por Mycoplasma/inmunología , Mycoplasma gallisepticum , Enfermedades de las Aves de Corral/genética , Enfermedades de las Aves de Corral/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Apoptosis , Ciclo Celular , Línea Celular , Proliferación Celular , Embrión de Pollo , Pollos , Citocinas/inmunología , Proteínas Quinasas Activadas por Mitógenos/inmunología , FN-kappa B/inmunología , Transducción de Señal , Regulación hacia Arriba
7.
J Cell Mol Med ; 24(11): 6178-6190, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32307881

RESUMEN

Mycoplasma gallisepticum (MG) can cause chronic respiratory disease (CRD) in chickens. While several studies have reported the inflammatory functions of microRNAs during MG infection, the mechanism by which exosomal miRNAs regulate MG-induced inflammation remains to be elucidated. The expression of exosome-microRNA derived from MG-infected chicken type II pneumocytes (CP-II) was screened, and the target genes and function of differentially expressed miRNAs (DEGs) were predicted. To verify the role of exosomal gga-miR-451, Western blot, ELISA and RT-qPCR were used in this study. The results showed that a total of 722 miRNAs were identified from the two exosomal small RNA (sRNA) libraries, and 30 miRNAs (9 up-regulated and 21 down-regulated) were significantly differentially expressed. The target miRNAs were significantly enriched in the treatment group, such as cell cycle, Toll-like receptor signalling pathway and MAPK signalling pathway. The results have also confirmed that gga-miR-451-absent exosomes derived from MG-infected CP-II cells increased inflammatory cytokine production in chicken fibroblast cells (DF-1), and wild-type CP-II cell-derived exosomes displayed protective effects. Collectively, our work suggests that exosomes from MG-infected CP-II cells alter the dynamics of the DF-1 cells, and may contribute to pathology of the MG infection via exosomal gga-miR-451 targeting YWHAZ involving in inflammation.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Exosomas/genética , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Inflamación/genética , MicroARNs/genética , Proteínas 14-3-3/metabolismo , Células Epiteliales Alveolares/ultraestructura , Animales , Apoptosis/genética , Ciclo Celular/genética , Línea Celular , Pollos/genética , Análisis por Conglomerados , Citocinas/metabolismo , Exosomas/metabolismo , Exosomas/ultraestructura , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Mediadores de Inflamación/metabolismo , MicroARNs/metabolismo , Anotación de Secuencia Molecular , Reproducibilidad de los Resultados , Receptores Toll-Like/metabolismo
8.
Microb Pathog ; 149: 104552, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33010363

RESUMEN

Mycoplasma gallisepticum (MG) infection is the main cause of chronic respiratory disease (CRD) characterized by severe respiratory inflammation in chickens. Polydatin (PD) is a resveratrol glycoside isolated from Polygonum cuspidatum, which has prominent anti-inflammatory effect. The purpose of this study was to investigate the therapeutic effect of PD against MG-induced inflammation in chicken and its underlying mechanism. Histopathological analysis showed that PD treatment (15, 30, and 45 mg/kg) apparently alleviated MG-induced pathological changes of chicken embryonic lung. In chicken embryo fibroblast (DF-1) cells, PD treatment (15, 30, and 60 µg/mL) could effectively suppress MG propagation, promote MG-infected cell proliferation and cell cycle progress, and inhibit MG-induced cell apoptosis. ELISA and qPCR assays showed that PD treatment significantly suppressed the expression of interleukin-6 (IL-6), IL-1ß and tumor necrosis factor-α (TNF-α) induced by MG both in vivo and in vitro. Besides, molecular studies indicated that the MG-induced levels of toll-like receptor-6 TLR6, myeloid differentiation-88 (MyD88) and nuclear factor κB (NF-κB) were significantly decreased by PD treatment. Moreover, immunofluorescence analysis showed that PD treatment restrained the MG-induced NF-κB-p65 nuclear translocation. Taken together, these results indicate the protective effects of PD against MG-induced inflammation injury in chicken were mainly by inhibiting the TLR6/MyD88/NF-κB pathway.


Asunto(s)
Mycoplasma gallisepticum , Animales , Embrión de Pollo , Pollos/metabolismo , Glucósidos , Inflamación/tratamiento farmacológico , Mycoplasma gallisepticum/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Estilbenos , Receptor Toll-Like 6/metabolismo , Factor de Necrosis Tumoral alfa
9.
Int J Mol Sci ; 20(12)2019 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-31238581

RESUMEN

MicroRNAs (miRNAs) have been determined to be important regulators for pathogenic microorganism infection. However, it is largely unclear how miRNAs are triggered during pathogen infection. We previously reported that the up-regulation of gga-miR-451 negatively regulates the Mycoplasma gallisepticum (MG)-induced production of inflammatory cytokines via targeting tyrosine3-monooxygenase/tryptophan5-monooxygenase activation protein zeta (YWHAZ). The aim of this study was to investigate the mechanism regulating gga-miR-451 in MG infection in chickens. Analysis of gga-miR-451 precursor, pri-miR-451, and pre-miR-451 indicated that the regulation occurred transcriptionally. We also identified the transcriptional regulatory region of gga-miR-451 that contained consensus-binding motif for aryl hydrocarbon receptor (AhR) and aryl hydrocarbon receptor nuclear translocator (Arnt) complex, which is known as the transcription factor that regulates gene expression. Luciferase reporter assays combined with chromatin immunoprecipitation (ChIP) demonstrated that AhR:Arnt bound directly to the promoter elements of gga-miR-451, which were responsible for gga-miR-451 transcription in the context of MG infection. Furthermore, upregulation of AhR:Arnt significantly induced gga-miR-451 and inhibited YWHAZ expression, suggesting that AhR:Arnt may play an anti-inflammatory role in MG infection. This discovery suggests that induced gga-miR-451 expression is modulated by AhR:Arnt in response to MG infection.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Interacciones Huésped-Patógeno/genética , MicroARNs/genética , Infecciones por Mycoplasma/genética , Infecciones por Mycoplasma/metabolismo , Mycoplasma gallisepticum , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Embrión de Pollo , Fibroblastos , Regulación de la Expresión Génica , Infecciones por Mycoplasma/microbiología , Activación Transcripcional
10.
Int J Mol Sci ; 20(5)2019 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-30818821

RESUMEN

Mycoplasma gallisepticum (MG) mainly infects chickens to initiate chronic respiratory disease (CRD). microRNAs (miRNAs) play vital roles according to previously reported studies. Our previous study showed that gga-miR-16-5p, in MG-infected lungs of chicken embryo, was upregulated by Illumina sequencing. The study aimed to reveal what role gga-miR-16-5p plays in CRD progression. gga-miR-16-5p was upregulated in MG-infected fibroblast cells (DF-1). Phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1) was demonstrated as the target gene of gga-miR-16-5p. Furthermore, PIK3R1 expression was lower in MG-infected groups than it in noninfected controls measured by qPCR. Additionally, overexpressed gga-miR-16-5p could downregulate PIK3R1 and phosphorylated serine/threonine kinase (p-Akt) to express protein, whereas there is an opposite effect on inhibition. Overexpressed gga-miR-16-5p resulted in decreased activity of tumor necrosis factor alpha (TNF-α) and the nuclear factor-kappaB (NF-κB) by qPCR. Furthermore, overexpressed gga-miR-16-5p restricted cell multiplication, cycle progression, and increased apoptosis of MG-infected DF-1 cells, whereas inhibited gga-miR-16-5p led to the opposite effect. Collectively, upregulated gga-miR-16-5p could decrease multiplication, cycle progression, and increase apoptosis of MG-infected DF-1 cells, at least partly through directly targeting PIK3R1 and inhibiting PI3K/Akt/NF-κB pathway to exert an anti-inflammatory effect. Our results will provide more experimental evidence to bring pathogenesis of MG infection to light.


Asunto(s)
Antiinflamatorios/metabolismo , Apoptosis/genética , Fibroblastos/metabolismo , Fibroblastos/microbiología , MicroARNs/genética , Mycoplasma gallisepticum/fisiología , Transducción de Señal , Regulación hacia Arriba/genética , Animales , Secuencia de Bases , Línea Celular , Proliferación Celular , Embrión de Pollo , Regulación hacia Abajo/genética , Pulmón/microbiología , Pulmón/patología , MicroARNs/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Enfermedades Respiratorias/genética , Enfermedades Respiratorias/microbiología , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
11.
Int J Mol Sci ; 19(4)2018 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-29652844

RESUMEN

Mycoplasma gallisepticum (MG) is the most economically significant mycoplasma pathogen of poultry that causes chronic respiratory disease (CRD) in chickens. Although miRNAs have been identified as a major regulator effect on inflammatory response, it is largely unclear how they regulate MG-induced inflammation. The aim of this study was to investigate the functional roles of gga-miR-451 and identify downstream targets regulated by gga-miR-451 in MG infection of chicken. We found that the expression of gga-miR-451 was significantly up-regulated during MG infection of chicken embryo fibroblast cells (DF-1) and chicken embryonic lungs. Overexpression of gga-miR-451 decreased the MG-induced inflammatory cytokine production, including tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and interleukin-6 (IL-6), whereas inhibition of gga-miR-451 had the opposite effect. Gene expression data combined with luciferase reporter assays demonstrated that tyrosine3-monooxygenase/tryptophan5-monooxygenase activation protein zeta (YWHAZ) was identified as a direct target of gga-miR-451 in the context of MG infection. Furthermore, upregulation of gga-miR-451 significantly inhibited the MG-infected DF-1 cells proliferation, induced cell-cycle arrest, and promoted apoptosis. Collectively, our results demonstrate that gga-miR-451 negatively regulates the MG-induced production of inflammatory cytokines via targeting YWHAZ, inhibits the cell cycle progression and cell proliferation, and promotes cell apoptosis. This study provides a better understanding of the molecular mechanisms of MG infection.


Asunto(s)
Proteínas 14-3-3/genética , Infecciones por Mycoplasma/genética , Mycoplasma gallisepticum/patogenicidad , Enfermedades de las Aves de Corral/microbiología , Infecciones del Sistema Respiratorio/veterinaria , Animales , Apoptosis , Línea Celular , Embrión de Pollo , Pollos , Citocinas/genética , Fibroblastos/química , Fibroblastos/citología , Fibroblastos/microbiología , Pulmón/química , Pulmón/microbiología , Infecciones por Mycoplasma/microbiología , Infecciones por Mycoplasma/veterinaria , Enfermedades de las Aves de Corral/genética , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/microbiología , Regulación hacia Arriba
12.
Int J Mol Sci ; 19(8)2018 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-30044397

RESUMEN

Mycoplasma gallisepticum (MG) is the pathogen of chronic respiratory disease (CRD), hallmarked by vigorous inflammation in chickens, causing the poultry industry enormous losses. miRNAs have emerged as important regulators of animal diseases. Previous miRNA sequencing data has demonstrated that miR-130b-3p is up-regulated in MG-infected chicken embryo lungs. Therefore, we aimed to investigate the function of miR-130b-3p in MG infection of chickens. RT-qPCR results confirmed that miR-130b-3p was up-regulated both in MG-infected chicken embryo lungs and chicken embryonic fibroblast cells (DF-1 cells). Furthermore, functional studies showed that overexpression of miR-130b-3p promoted MG-infected DF-1 cell proliferation and cell cycle, whereas inhibition of miR-130b-3p weakened these cellular processes. Luciferase reporter assay combined with gene expression data supported that phosphatase and tensin homolog deleted on chromosome ten (PTEN) was a direct target of miR-130b-3p. Additionally, overexpression of miR-130b-3p resulted in up-regulations of phosphatidylinositol-3 kinase (PI3K), serine/threonine kinase (AKT), and nuclear factor-κB (NF-κB), whereas inhibition of miR-130b-3p led to the opposite results. Altogether, upon MG infection, up-regulation of miR-130b-3p activates the PI3K/AKT/NF-κB pathway, facilitates cell proliferation and cell cycle via down-regulating PTEN. This study helps to understand the mechanism of host response to MG infection.


Asunto(s)
Pollos/microbiología , MicroARNs/metabolismo , Infecciones por Mycoplasma/veterinaria , Mycoplasma gallisepticum/patogenicidad , Enfermedades de las Aves de Corral/microbiología , Animales , Ciclo Celular , Línea Celular , Proliferación Celular , Embrión de Pollo , Fibroblastos/microbiología , Humanos , Pulmón/microbiología , MicroARNs/genética , Infecciones por Mycoplasma/microbiología , FN-kappa B/genética , FN-kappa B/metabolismo , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Enfermedades de las Aves de Corral/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Regulación hacia Arriba
13.
J Cell Mol Med ; 21(11): 2796-2808, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28524642

RESUMEN

Staphylococcus aureus (S. aureus) causes severe inflammation in various infectious diseases, leading to high mortality. The clinical application of antibiotics has gained a significant curative effect. However, it has led to the emergence of various resistant bacteria. Therefore, in this study, we investigated the protective effect of polydatin (PD), a traditional Chinese medicine extract, on S. aureus lipoteichoic acid (LTA)-induced injury in vitro and in vivo. First, a significant improvement in the pathological conditions of PD in vivo was observed, suggesting that PD had a certain protective effect on LTA-induced injury in a mouse model. To further explore the underlying mechanisms of this protective effect of PD, LTA-induced murine macrophages were used in this study. The results have shown that PD could reduce the NF-κB p65, and IκBα phosphorylation levels increased by LTA, resulting in a decrease in the transcription of pro-inflammatory factors, such as TNF-α, IL-1ß and IL-6. However, LTA can not only activate NF-κB through the recognition of TLR2 but also increase the level of intracellular reactive oxygen species (ROS), thereby activating NF-κB signalling. We also detected high levels of ROS that activate caspases 9 and 3 to induce apoptosis. In addition, using a specific NF-κB inhibitor that could attenuate apoptosis, namely NF-κB p65, acted as a pro-apoptotic transcription factor in LTA-induced murine macrophages. However, PD could inhibit the generation of ROS and NF-κB p65 activation, suggesting that PD suppressed LTA-induced injury by attenuating ROS generation and TLR2-NFκB signalling.


Asunto(s)
Antioxidantes/farmacología , Endometriosis/tratamiento farmacológico , Glucósidos/farmacología , Sustancias Protectoras/farmacología , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Estilbenos/farmacología , Receptor Toll-Like 2/genética , Factor de Transcripción ReIA/genética , Animales , Supervivencia Celular/efectos de los fármacos , Endometriosis/inducido químicamente , Endometriosis/genética , Endometriosis/inmunología , Femenino , Regulación de la Expresión Génica , Proteínas I-kappa B/genética , Proteínas I-kappa B/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/aislamiento & purificación , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos BALB C , Estrés Oxidativo/efectos de los fármacos , Células RAW 264.7 , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Staphylococcus aureus/química , Ácidos Teicoicos/antagonistas & inhibidores , Ácidos Teicoicos/aislamiento & purificación , Ácidos Teicoicos/toxicidad , Receptor Toll-Like 2/inmunología , Factor de Transcripción ReIA/inmunología , Útero/efectos de los fármacos , Útero/inmunología , Útero/patología
14.
Acta Pharmacol Sin ; 38(2): 211-222, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27890916

RESUMEN

Recent studies show that Polydatin (PD) extracted from the roots of Polygonum cuspidatum Sieb, a widely used traditional Chinese remedies, possesses anti-inflammatory activity in several experimental models. In this study, we investigated the anti-inflammatory effects of PD on Staphylococcus aureus-induced mastitis in mice and elucidated the potential mechanisms. In mice with S aureus-induced mastitis, administration of PD (15, 30, 45 mg/kg, ip) or dexamethasone (Dex, 5 mg/kg, ip) significantly suppressed the infiltration of inflammatory cells, ameliorated the mammary structural damage, and inhibited the activity of myeloperoxidase, a biomarker of neutrophils accumulation. Furthermore, PD treatment dose-dependently decreased the levels of TNF-α, IL-1ß, IL-6 and IL-8 in the mammary gland tissues. PD treatment also dose-dependently decreased the expression of TLR2, MyD88, IRAK1, IRAK4 and TRAF6 as well as the phosphorylation of TAK1, MKK3/6, p38 MAPK, IκB-α and NF-κB in the mammary gland tissues. In mouse mammary epithelial cells (mMECs) infected by S aureus in vitro, pretreatment with PD dose-dependently suppressed the upregulated pro-inflammatory cytokines and signaling proteins, and the nuclear translocation of NF-κB p65 and AP-1. A TLR2-neutralizing antibody mimicked PD in its suppression on S aureus-induced upregulation of MyD88, p-p38 and p-p65 levels in mMECs. PD (50, 100 µg/mL) affected neither the growth of S aureus in vitro, nor the viability of mMECs. In conclusion, PD does not exhibit antibacterial activity against S aureus, its therapeutic effects in mouse S aureus-induced mastitis depend on its ability to down-regulate pro-inflammatory cytokine levels via inhibiting TLR2-mediated activation of the p38 MAPK/NF-κB signaling pathway.


Asunto(s)
Glucósidos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mastitis/tratamiento farmacológico , FN-kappa B/metabolismo , Staphylococcus aureus/efectos de los fármacos , Estilbenos/farmacología , Receptor Toll-Like 2/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Antiinflamatorios/farmacología , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Dexametasona/farmacología , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Femenino , Mediadores de Inflamación/metabolismo , Mastitis/inmunología , Mastitis/metabolismo , Mastitis/microbiología , Ratones , Infiltración Neutrófila/efectos de los fármacos , Peroxidasa/antagonistas & inhibidores , Staphylococcus aureus/inmunología , Receptor Toll-Like 2/inmunología
15.
Appl Microbiol Biotechnol ; 100(3): 1343-1354, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26549235

RESUMEN

The adhesin protein from Mycoplasma gallisepticum (HS strain), namely pMGA1.2, is required for M. gallisepticum (MG) infection in chicken. However, the host factor(s) that interact with pMGA1.2 is not known. In this study, we prepared the membrane fraction of trachea epithelial cells from chicken embryos. Using an improved virus overlay protein blot assay (VOPBA) and glutathione S-transferase (GST) pull-down assay, we found that pMGA1.2 specifically bound to a ∼30 kDa host protein. This host protein was further identified by mass spectrometry as chicken apolipoprotein A-I (ApoA-I). We expressed and purified the recombinant ApoA-I protein in Escherichia coli and confirmed that it bound to the purified pMGA1.2 protein in vitro. Transiently expressed pMGA1.2 and ApoA-I were colocalized in HeLa cells. Finally, we designed small interfering RNA (siRNA) molecules to knock down the expression of either ApoA-I or pMGA1.2, which inhibited the MG-induced cell cycle disruption in cells of chicken embryo fibroblast cell line (DF-1). Similarly, knockdown of ApoA-I inhibited the cilia loss and damage in chicken trachea cells in MG infection. In summary, ApoA-I may be an essential host factor in MG infection through interacting with pMGA1.2.


Asunto(s)
Apolipoproteína A-I/metabolismo , Proteínas Aviares/metabolismo , Proteínas Bacterianas/metabolismo , Lipoproteínas/metabolismo , Infecciones por Mycoplasma/veterinaria , Mycoplasma gallisepticum/metabolismo , Enfermedades de las Aves de Corral/metabolismo , Animales , Apolipoproteína A-I/genética , Proteínas Aviares/genética , Proteínas Bacterianas/genética , Pollos , Interacciones Huésped-Patógeno , Lipoproteínas/genética , Infecciones por Mycoplasma/genética , Infecciones por Mycoplasma/metabolismo , Infecciones por Mycoplasma/microbiología , Mycoplasma gallisepticum/genética , Enfermedades de las Aves de Corral/genética , Enfermedades de las Aves de Corral/microbiología , Unión Proteica
16.
Int J Mol Sci ; 16(12): 28669-82, 2015 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-26633386

RESUMEN

Mycoplasma gallisepticum (MG), one of the most pathogenic Mycoplasma, has caused tremendous economic loss in the poultry industry. Recently, increasing evidence has suggested that micro ribonucleic acids (miRNAs) are involved in microbial pathogenesis. However, little is known about potential roles of miRNAs in MG infection of chicken. In the present study, using miRNA Solexa sequencing we have found that gga-miR-101-3p was up-regulated in the lungs of MG-infected chicken embryos. Moreover, gga-miR-101-3p regulated expression of the host enhancer of zeste homolog 2 (EZH2) through binding to the 3' un-translated region (3'-UTR) of EZH2 gene. Over-expression of gga-miR-101-3p significantly inhibited EZH2 expression and hence inhibited proliferation of chicken embryonic fibroblast (DF-1 cells) by blocking the G1-to-S phase transition. Similar results were obtained in MG-infected chicken embryos and DF-1 cells, where gga-miR-101-3p was significantly up-regulated, while EZH2 was significantly down-regulated. This study reveals that gga-miR-101-3p plays an important role in MG infection through regulation of EZH2 expression and provides a new insight into the mechanisms of MG pathogenesis.


Asunto(s)
MicroARNs/genética , Infecciones por Mycoplasma/veterinaria , Mycoplasma gallisepticum , Enfermedades de las Aves de Corral/genética , Enfermedades de las Aves de Corral/microbiología , Regiones no Traducidas 3' , Animales , Secuencia de Bases , Sitios de Unión , Ciclo Celular , Proliferación Celular , Embrión de Pollo , Pollos , Regulación de la Expresión Génica , MicroARNs/química , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Interferencia de ARN , ARN Mensajero/química , ARN Mensajero/genética , Alineación de Secuencia
17.
Poult Sci ; 103(5): 103652, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38537405

RESUMEN

Mycoplasma gallisepticum (MG) is a highly contagious avian respiratory pathogen characterized by rapid spread, widespread distribution, and long-term persistence of infection. Previous studies have shown that chicken macrophage HD11 cells play a critical role in the replication and immunomodulation of MG. Macrophages are multifunctional immunomodulatory cells that polarize into different functions and morphologies in response to exogenous stimuli. However, the effect of MG infection on HD11 polarization is not well understood. In this study, we observed a time-dependent increase in both the expression of the MG-related virulence protein pMGA1.2 and the copy number of MG upon MG infection. Polarization studies revealed an upregulation of M1-type marker genes in MG-infected HD11 cells, suggesting that MG mainly induces HD11 macrophages towards M1-type polarization. Furthermore, MG activated the inflammatory vesicle NLRP3 signaling pathway, and NLRP3 inhibitors affected the expression of M1 and M2 marker genes, indicating the crucial regulatory role of the NLRP3 signaling pathway in MG-induced polarization of HD11 macrophages. Our findings reveal a novel mechanism of MG infection, namely the polarization of MG-infected HD11 macrophages. This discovery suggests that altering the macrophage phenotype to inhibit MG infection may be an effective control strategy. These findings provide new perspectives on the pathogenic mechanism and control measures of MG.


Asunto(s)
Pollos , Macrófagos , Infecciones por Mycoplasma , Mycoplasma gallisepticum , Enfermedades de las Aves de Corral , Mycoplasma gallisepticum/fisiología , Animales , Macrófagos/inmunología , Macrófagos/microbiología , Enfermedades de las Aves de Corral/microbiología , Enfermedades de las Aves de Corral/inmunología , Infecciones por Mycoplasma/veterinaria , Infecciones por Mycoplasma/inmunología , Infecciones por Mycoplasma/microbiología , Línea Celular
18.
Vet Microbiol ; 296: 110175, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39018941

RESUMEN

Mycoplasma gallisepticum (MG) is the primary causative agent of chronic respiratory disease (CRD) in chickens, characterized by respiratory inflammation. S100A9 plays a pivotal role in modulating the inflammatory response to microbial pathogens. Our prior investigation revealed a significant upregulation of S100A9 in the lungs of chickens following MG infection. This study delves into the immunomodulatory effects of S100A9 during MG infection, demonstrating a notable increase in S100A9 levels in the lungs, immune organs, alveolar epithelial type II cells (AECII), and macrophage HD11 cells of MG-infected chicks and embryos. In MG-infected AECII cells, S100A9 overexpression significantly enhanced MG proliferation and adhesion, suppressed AVBD1, NFκB, pro-inflammatory factors (IL1ß and TNFα), and chemokines, reduced apoptosis, and promoted cell proliferation, thereby facilitating MG infection. Conversely, inhibiting S100A9 produced opposing effects. In MG-infected HD11 cells, S100A9 impeded MG proliferation and adhesion, increased AVBD1, NFκB, pro-inflammatory factors, and chemokines, and induced cell apoptosis while inhibiting proliferation. Additional results demonstrated that S100A9 facilitates MG infection by modulating the TLR7/NFκB/JAK/STAT pathway in AECII/HD11 cells. In summary, S100A9 exhibits a dual role in activating/inhibiting the natural immune response through TLR7/NFκB/JAK/STAT pathway regulation. This dual role promotes MG infection in AECII cells while enabling MG to evade immune surveillance by HD11 cells, ultimately enhancing the overall infection process. These findings advance our understanding of host-pathogen interactions during MG infection and underscore S100A9's potential as a therapeutic target for CRD in chickens.


Asunto(s)
Calgranulina B , Pollos , Infecciones por Mycoplasma , Mycoplasma gallisepticum , Enfermedades de las Aves de Corral , Animales , Mycoplasma gallisepticum/inmunología , Infecciones por Mycoplasma/veterinaria , Infecciones por Mycoplasma/inmunología , Infecciones por Mycoplasma/microbiología , Pollos/inmunología , Enfermedades de las Aves de Corral/microbiología , Enfermedades de las Aves de Corral/inmunología , Calgranulina B/genética , Calgranulina B/metabolismo , Línea Celular , Pulmón/microbiología , Pulmón/inmunología , Embrión de Pollo , FN-kappa B/metabolismo , Proliferación Celular , Macrófagos/inmunología , Macrófagos/microbiología
19.
Anim Genet ; 44(2): 214-6, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22591010

RESUMEN

In this study, we performed a sequence characterization of the duck melanocortin 1 receptor (alpha-melanocyte stimulating hormone receptor) (MC1R) gene to analyze the relationship between MC1R polymorphism and the extended black variant in domestic ducks based on the extended black (E) and non-extended black (e(+) ) allele hypothesis of the duck MC1R gene. Both c.52G>A and c.376G>A substitutions are highly associated with the duck extended black variant (P < 0.01), but the novel c.52G>A substitution is more of a fit for the allele hypothesis of the duck MC1R gene.


Asunto(s)
Patos/genética , Fenotipo , Pigmentación/genética , Polimorfismo de Nucleótido Simple/genética , Receptor de Melanocortina Tipo 1/genética , Animales , Secuencia de Bases , Patos/fisiología , Frecuencia de los Genes , Estudios de Asociación Genética , Modelos Genéticos , Datos de Secuencia Molecular , Pigmentación/fisiología , Análisis de Secuencia de ADN/veterinaria
20.
Cells ; 12(19)2023 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-37830635

RESUMEN

Respiratory diseases represent a significant economic and health burden worldwide, affecting millions of individuals each year in both human and animal populations. MicroRNAs (miRNAs) play crucial roles in gene expression regulation and are involved in various physiological and pathological processes. Exosomal miRNAs and cellular miRNAs have been identified as key regulators of several immune respiratory diseases, such as chronic respiratory diseases (CRD) caused by Mycoplasma gallisepticum (MG), Mycoplasma pneumoniae pneumonia (MMP) caused by the bacterium Mycoplasma pneumoniae, coronavirus disease 2019 (COVID-19), chronic obstructive pulmonary disease (COPD), asthma, and acute lung injury/acute respiratory distress syndrome (ALI/ARDS). Consequently, miRNAs seem to have the potential to serve as diagnostic biomarkers and therapeutic targets in respiratory diseases. In this review, we summarize the current understanding of the functional roles of miRNAs in the above several respiratory diseases and discuss the potential use of miRNAs as stable diagnostic biomarkers and therapeutic targets for several immune respiratory diseases, focusing on the identification of differentially expressed miRNAs and their targeting of various signaling pathways implicated in disease pathogenesis. Despite the progress made, unanswered questions and future research directions are discussed to facilitate personalized and targeted therapies for patients with these debilitating conditions.


Asunto(s)
COVID-19 , MicroARNs , Mycoplasma gallisepticum , Enfermedad Pulmonar Obstructiva Crónica , Síndrome de Dificultad Respiratoria , Animales , Humanos , MicroARNs/genética , MicroARNs/metabolismo , COVID-19/genética , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Síndrome de Dificultad Respiratoria/genética , Biomarcadores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA