Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Theriogenology ; 214: 307-313, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-37956579

RESUMO

Endometritis is one of the important reasons for the low fecundity of dairy cows, which has brought huge economic losses to the dairy industry. Emerging evidence suggests that miR-92b is a novel therapeutic molecule that plays a crucial role in many inflammatory diseases. However, its mechanism in lipoteichoic acid (LTA) induced endometritis remains unclear. In the present study, we explored the mechanism of miR-92b on LTA-induced endometritis in vivo and in vitro. The result displayed that the expression of miR-92b was reduced in LTA induced mouse endometritis and bovine endometrial epithelial cell lines (BEND). Overexpression miR-92b significantly alleviated mouse uterine injury and reduced the protein levels of TNF-α, IL-1ß and the MPO activity. The reporter assay of luciferase showed that miR-92b directly targeted the transmembrane receptor Frizzled-10 (FZD10), a transmembrane-type Wnt receptor. Molecular experiments were further performed to explore the mechanism of miR-92b in protecting LTA induced endometritis. The results of in vitro suggested that miR-92b mimic decreased the protein levels of Wnt3a and ß-catenin in LTA stimulated BEND, which were abolished by overexpression of FZD10. As expected, miR-92b mimic decreased the expression levels of TNF-α and IL-1ß, while overexpression of FZD10 promoted the production of these pro-inflammatory cytokines. Collectively, the above findings indicated that miR-92b might be an effective strategy for treatment of LTA induced endometritis.


Assuntos
Doenças dos Bovinos , Endometrite , MicroRNAs , Feminino , Animais , Bovinos , Camundongos , Endometrite/veterinária , MicroRNAs/genética , MicroRNAs/metabolismo , Fator de Necrose Tumoral alfa/genética , beta Catenina/genética
2.
Microbiol Spectr ; 11(3): e0040923, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37067455

RESUMO

Endometritis is a common cow disease characterized by inflammation of endometrium, which leads to infertility or low fertility of cows and brings huge economic losses to the dairy industry. Tau interferon (IFN-τ) has many important biological functions, including an anti-inflammatory effect. The present study aimed to survey the effects of IFN-τ administration on gut microflora and body metabolism in mice with endometritis and to explore the potential relationship. The results indicated that IFN-τ obviously alleviated the damage and ultrastructural changes of mouse endometrium induced by Escherichia coli and enhanced tight junction protein's expression level. Through analysis by 16S rRNA gene sequencing, we found that IFN-τ, especially at 12 h, could regulate the composition of gut microbiota associated with Pediococcus, Staphylococcus, and Enterorhabdus in E. coli-induced mouse endometritis. Through histometabonomics, it was found that endometritis was related to 11 different metabolites and 4 potential metabolic pathways. These metabolites and metabolic pathways were major participants in metabolic pathways, cysteine and methionine metabolism, arachidonic acid metabolism, and pyrimidine metabolism. Correlation analysis of gut microbiota with uterine tissue metabolomics showed that changes in metabolic pathways might be affected by gut microbiota, such as Enterorhabdus in mouse endometritis. The above results indicated that the anti-inflammatory mechanism of IFN-τ might be reduction of the abundance of Enterorhabdus in the gut microbiota, affecting the expression level of important metabolites in uterine tissue and thus playing an anti-inflammatory role. IMPORTANCE The change in intestinal flora has been the focus of many disease studies in recent years, but the pathogenetic effect of interferon on endometritis is still unclear. The results of this study showed that IFN-τ alleviated the damage in mouse endometritis induced by E. coli and improved the endometrial tissue barrier. Its functional mechanism may be reduction of the abundance of Enterorhabdus in the intestinal microbiota, affecting the expression level of important metabolites in uterine tissue and thus playing an anti-inflammatory role.


Assuntos
Endometrite , Microbioma Gastrointestinal , Humanos , Feminino , Animais , Camundongos , Bovinos , Endometrite/tratamento farmacológico , Endometrite/genética , Endometrite/veterinária , RNA Ribossômico 16S/genética , Escherichia coli/genética , Genes de RNAr , Metabolômica , Anti-Inflamatórios/farmacologia
3.
J Inflamm Res ; 14: 2963-2978, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34262322

RESUMO

PURPOSE: Fisetin is a natural flavone of polyphenol, which widely exists in many fruits and vegetables and has many pharmacological activities. However, the mechanism involved remains largely unknown. Here, we investigate the mechanisms of fisetin on the inflammatory response and oxidative stress in LPS-induced endometritis model and bovine endometrial epithelial cell line (BEND). METHODS: The function of fisetin was analyzed by network pharmacology. Effects of increasing doses of fisetin on inflammation and oxidative stress are studied in BALB/c mice with LPS-induced endometritis. The underlying mechanisms of antioxidant activity of fisetin were further explored in LPS-stimulated BEND cells. RESULTS: The results showed that fisetin significantly alleviated LPS-induced inflammatory injury and oxidative stress both in vivo and in vitro. Further studies found that fisetin greatly inhibited the LPS stimulated TLR4 expression and nuclear translocation of nuclear factor-κB (NF-κB), thus reducing the pro-inflammatory mediators secretion. Silencing TLR4 reduced LPS-induced inflammatory responses. Moreover, we observed that fisetin evidently decreased ROS production but activated Nrf2/HO-1 pathway in LPS-stimulated BEND cells. To further explore the role of Nrf2 in fisetin-induced HO-1 protein expression, the specific siRNA was used to silence Nrf2 expression. Silencing Nrf2 abrogated the inhibitory effects of fisetin on LPS-induced pro-inflammatory cytokines TNF-α, IL-1ß secretion, NADPH oxidase-4 (Nox4) and ROS production. CONCLUSION: In conclusion, fisetin effectively protected against LPS-induced oxidative stress and inflammatory responses which may be closely correlated to inhibition of TLR4-mediated ROS/NF-κB and activation of the Nrf2/HO-1 pathway.

5.
J Cell Mol Med ; 24(1): 405-417, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31756048

RESUMO

Endometritis is a postnatal reproductive disorder disease, which leads to great economic losses for the modern dairy industry. Emerging evidence indicates that microRNAs (miRNAs) play a pivotal role in a variety of diseases and have been identified as critical regulators of the innate immune response. Recent miRNome profile analysis revealed an altered expression level of miR-148a in cows with endometritis. Therefore, the present study aims to investigate the regulatory role of miR-148a in the innate immune response involved in endometritis and estimate its potential therapeutic value. Here, we found that miR-148a expression in lipopolysaccharide (LPS)-stimulated endometrial epithelial cells was significantly decreased. Our results also showed that overexpression of miR-148a using agomiR markedly reduced the production of pro-inflammatory cytokines, such as IL-1ß and TNF-α. Moreover, overexpression of miR-148a also suppressed NF-κB p65 activation by targeting the TLR4-mediated pathway. Subsequently, we further verified that miR-148a repressed TLR4 expression by binding to the 3'-UTR of TLR4 mRNA. Additionally, an experimental mouse endometritis model was employed to evaluate the therapeutic value of miR-148a. In vivo studies suggested that up-regulation of miR-148a alleviated the inflammatory conditions in the uterus as evidenced by H&E staining, qPCR and Western blot assays, while inhibition of miR-148a had inverse effects. Collectively, pharmacologic stabilization of miR-148a represents a novel therapy for endometritis and other inflammation-related diseases.


Assuntos
Endometrite/genética , Inflamação/genética , MicroRNAs/metabolismo , Animais , Sequência de Bases , Bovinos , Citocinas/biossíntese , Endometrite/patologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos , Camundongos Endogâmicos BALB C , MicroRNAs/genética , NF-kappa B/metabolismo , Transdução de Sinais
6.
Mol Ther ; 27(10): 1758-1771, 2019 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-31405809

RESUMO

Emerging evidence has revealed that excessive activation of macrophages may result in an adverse lung inflammation involved in sepsis-related acute lung injury (ALI). However, it has never been clearly identified whether peripheral circulating serum exosomes participate in the pathogenesis of sepsis-related ALI. Therefore, the purposes of our study were to investigate the effect of serum exosomes on macrophage activation and elucidate a novel mechanism underlying sepsis-related ALI. Here we found that exosomes were abundant in the peripheral blood from ALI mice and selectively loaded microRNAs (miRNAs), such as miR-155. In vivo experiments revealed that intravenous injection of serum exosomes harvested from ALI mice, but not control mice, increased the number of M1 macrophages in the lung, and it caused lung inflammation in naive mice. In vitro, we demonstrated that serum exosomes from ALI mice delivered miR-155 to macrophages, stimulated nuclear factor κB (NF-κB) activation, and induced the production of tumor necrosis factor alpha (TNF-α) and interleukin (IL)-6. Furthermore, we also showed that serum exosome-derived miR-155 promoted macrophage proliferation and inflammation by targeting SHIP1 and SOCS1, respectively. Collectively, our data suggest the important role of circulating exosomes secreted into peripheral blood as a key mediator of septic lung injury via exosome-shuttling miR-155.


Assuntos
Lesão Pulmonar Aguda/genética , Exossomos/transplante , MicroRNAs/genética , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases/genética , Pneumonia/genética , Proteína 1 Supressora da Sinalização de Citocina/genética , Regiões 3' não Traduzidas , Lesão Pulmonar Aguda/sangue , Animais , Modelos Animais de Doenças , Exossomos/genética , Injeções Intravenosas , Interleucina-6/metabolismo , Ativação de Macrófagos , Macrófagos Alveolares/imunologia , Masculino , Camundongos , NF-kappa B/metabolismo , Pneumonia/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
7.
Int Immunopharmacol ; 73: 41-48, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31078924

RESUMO

As a key virulence factor of Mycobacterium tuberculosis, EsxA is not only involved in phagosome rupture, but also functions in stimulation of immune responses in macrophages. Here, we report thatmiR-148a is down-regulated in the macrophages infected with Mycobacterium marinum (Mm). Using the knockout strain Mm∆EsxA/B, recombinant EsxA, EsxB and EsxA/B heterodimer proteins, we provide evidence that down-regulation of miR-148ais dependent on EsxA, and up-regulation of miR-148a reduces Mm intracellular survival. Moreover, up-regulation of miR-148a down-regulates the pro-inflammatory cytokines (e.g. TNF-α and IL-1ß) and the TLR4-mediated NF-κB activation. Together, miR-148a may function as an anti-inflammation modulator in responses to mycobacterial infection. Regulation of miR-148a may provide a novel venue in development of therapies in tuberculosis.


Assuntos
Proteínas de Bactérias/genética , MicroRNAs , Mycobacterium marinum/genética , Animais , Regulação para Baixo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Camundongos , NF-kappa B/metabolismo , Células RAW 264.7 , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
8.
Microb Pathog ; 132: 302-312, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31059756

RESUMO

Acute lung injury (ALI) is clinically characterized by excessive inflammation leading to acute respiratory distress syndrome (ARDS), having high morbidity and mortality both in human and animals. Ginsenoside Rb1 (Rb1) is a major primary bioactive component extracted by Panax ginseng, which has numerous pharmacological functions such as anti-cancer, anti-inflammatory, and antioxidant. However, the anti-inflammatory effects of Rb1 in Staphylococcus aureus (S. aureus)-induced ALI in mice have not been investigated. The aim of the current study was to determine the anti-inflammatory influence of Rb1 on S. aureus-induced ALI in mice, and to explore its possible underlying principle mechanisms in RAW 264.7 macrophage cells. The results of physical morphology, histopathological variation and wet-to-dry weight ratio of lungs revealed that Rb1 significantly attenuated S. aureus-induced lung injury. Furthermore, qPCR results displayed that Rb1 inhibited IL-1ß, IL-6 and TNF-α production both in vivo and in vitro. The activation of Toll-like receptor 2 (TLR2) by S. aureus was inhibited by application of Rb1 as confirmed by results of immunofluorescence assay. The expression of NF-kB and MAPK signaling proteins revealed that Rb1 significantly attenuated the phosphorylation of p65, ERK, as well as JNK. Altogether, the results of this experiment presented that Rb1 has ability to protect S. aureus-induced ALI in mice by attenuating TLR-2-mediated NF-kB and MAPK signaling pathways. Consequently, Rb-1 might be a potential medicine in the treatment of S. aureus-induced lung inflammation.


Assuntos
Lesão Pulmonar Aguda/microbiologia , Ginsenosídeos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , NF-kappa B/metabolismo , Staphylococcus aureus/efeitos dos fármacos , Animais , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Pulmão/patologia , Masculino , Camundongos , Panax/química , Pneumonia , Células RAW 264.7/efeitos dos fármacos , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
9.
Int Immunopharmacol ; 70: 201-207, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30822611

RESUMO

Endometritis is one of the main diseases that causes great economic losses in the dairy industry. Recent studies have shown that matrine extracted from the traditional Chinese herb Sophora flavescens is an alkaloid with a broad range of bioactivities. Here, we aimed to investigate the protective effects of matrine on Staphylococcus aureus lipoteichoic acid (LTA)-induced endometritis in mice and elucidate the possible molecular mechanisms in vitro. Histopathological changes showed that matrine remarkably attenuated the uterus injury in a mouse model of LTA-induced endometritis. qPCR and ELISA results showed that matrine dose-dependently reduced the expression of pro-inflammatory cytokines (TNF-α and IL-1ß). To further elucidate the underlying mechanisms of this protective effect of matrine, LTA-stimulated bovine endometrial epithelial cells (bEECs) were employed in this study. The results demonstrated that TLR2 expression and its downstream nuclear factor (NF)-κB activation were both suppressed by matrine treatment. Furthermore, a small interference RNA targeting TLR2 gene mimicked matrine in its inhibition on LTA-induced activation of TLR2 and NF-κB. In conclusion, these findings suggest the protective effect of matrine against LTA-induced endometritis through negative regulation of TLR2-mediated NF-κB pathway.


Assuntos
Alcaloides/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Endometrite/tratamento farmacológico , Medicina Tradicional Chinesa , Quinolizinas/uso terapêutico , Staphylococcus aureus/fisiologia , Útero/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Regulação para Baixo , Endometrite/induzido quimicamente , Endometrite/imunologia , Feminino , Humanos , Interleucina-1beta/metabolismo , Lipopolissacarídeos/metabolismo , Camundongos , NF-kappa B/metabolismo , RNA Interferente Pequeno/genética , Transdução de Sinais , Sophora/imunologia , Ácidos Teicoicos/metabolismo , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Útero/patologia , Matrinas
10.
J Cell Mol Med ; 23(5): 3711-3723, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30920152

RESUMO

It is well established that cancer cells depend upon aerobic glycolysis to provide the energy they need to survive and proliferate. However, anti-glycolytic agents have yielded few positive results in human patients, in part due to dose-limiting side effects. Here, we discovered the unexpected anti-cancer efficacy of Polydatin (PD) combined with 2-deoxy-D-glucose (2-DG), which is a compound that inhibits glycolysis. We demonstrated in two breast cell lines (MCF-7 and 4T1) that combination treatment with PD and 2-DG induced cell apoptosis and inhibited cell proliferation, migration and invasion. Furthermore, we determined the mechanism of PD in synergy with 2-DG, which decreased the intracellular reactive oxygen (ROS) levels and suppressed the PI3K/AKT pathway. In addition, the combined treatment inhibited the glycolytic phenotype through reducing the expression of HK2. HK2 deletion in breast cancer cells thus improved the anti-cancer activity of 2-DG. The combination treatment also resulted in significant tumour regression in the absence of significant morphologic changes in the heart, liver or kidney in vivo. In summary, our study demonstrates that PD synergised with 2-DG to enhance its anti-cancer efficacy by inhibiting the ROS/PI3K/AKT/HIF-1α/HK2 signalling axis, providing a potential anti-cancer strategy.


Assuntos
Neoplasias da Mama/metabolismo , Desoxiglucose/farmacologia , Enzimas/metabolismo , Glucosídeos/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Estilbenos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Desoxiglucose/química , Enzimas/genética , Feminino , Glucosídeos/química , Glicólise/efeitos dos fármacos , Hexoquinase/genética , Hexoquinase/metabolismo , Humanos , Células MCF-7 , Camundongos Endogâmicos BALB C , Estrutura Molecular , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estilbenos/química , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
11.
Mol Med Rep ; 19(3): 2279-2286, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30664199

RESUMO

Sodium houttuyfonate (SH) has been indicated to play an important anti­inflammatory role. Previous studies have confirmed that SH can inhibit the NF­κB pathway in lipopolysaccharide (LPS)­induced mastitis in bovine mammary epithelial cells. However, the effects of SH on LPS­induced mastitis in animals should be verified to further evaluate its actual value. In the present study, the anti­inflammatory effects of SH were investigated in mouse models and a mouse mammary epithelial cell line. Hematoxylin and eosin staining (H&E) showed that SH therapy significantly alleviated the pathological changes in mammary glands. Myeloperoxidase (MPO) activity analysis demonstrated that SH substantially decreased MPO activity in vivo. RT­qPCR results showed that SH reduced the expression of interleukin (IL)­1, IL­6 and tumor necrosis factor α both in vivo and in vitro. In addition, western blot results indicated that SH suppressed the phosphorylation of nuclear factor kappa­light­chain­enhancer of activated B­cells (NF­κB) p65 protein and reduced the degradation of inhibitor of kappa light polypeptide gene enhancer in B­cells alpha protein in vivo and in vitro. These results demonstrated that SH ameliorates LPS­induced mastitis by inhibiting the NF­κB pathway.


Assuntos
Alcanos/administração & dosagem , Inflamação/tratamento farmacológico , Mastite/tratamento farmacológico , Sulfitos/administração & dosagem , Fator de Transcrição RelA/genética , Animais , Bovinos , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/patologia , Lipopolissacarídeos/toxicidade , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/patologia , Mastite/induzido quimicamente , Mastite/genética , Mastite/patologia , Camundongos , NF-kappa B/genética , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética
12.
Reprod Sci ; 26(6): 829-838, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30526368

RESUMO

Previous studies show that methylseleninic acid (MSA), which is the most common selenium derivative used as a drug in humans, exerts specific cytotoxic effects in several cancer cell types. However, the complex mechanism of these effects has not been fully elucidated. Here, we demonstrate by Cell Counting Kit-8 in mouse breast cancer cell line 4T1 that MSA inhibits cell viability in a concentration-dependent (5, 10, 20 µmol/L) and time-dependent (6, 12, 24 hours) manner. Flow cytometry, Western blot, and Reverse Transcription-Polymerase Chain Reaction (RT-PCR) analyses indicated that MSA inhibits cancer cell invasion and induces apoptosis by the activation of caspase-3, poly ADP ribose polymerase 1 (PARP1), and BCL2-associated X. Furthermore, MSA demonstrated anticancer activity by inhibiting the Janus kinase 2/signal transducers and activators of transcription 3 (JAK2/STAT3) pathway. The MSA treatment for 24 hours decreased the phosphorylation of JAK2 and STAT3 in 4T1 cells by Western blot. We also confirmed this with the use of a JAK2 chemical inhibitor, AG490, as a positive control. In a 4T1 orthotopic allograft model, morphological and TdT-mediated dUTP nick-end labeling analyses showed that MSA treatment (1.5 mg/kg/weight) for 28 days inhibits tumor growth consistent with the clinical anticancer drug cyclophosphamide. Our observations demonstrate that MSA is a potent anticancer drug in breast cancer and uncovered a key role of the JAK2/STAT3 pathway in modulating tumor growth.


Assuntos
Antineoplásicos , Neoplasias da Mama/tratamento farmacológico , Janus Quinase 2/antagonistas & inibidores , Compostos Organosselênicos/uso terapêutico , Fator de Transcrição STAT3/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Janus Quinase 2/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Compostos Organosselênicos/farmacologia , Fator de Transcrição STAT3/fisiologia
13.
J Cell Physiol ; 234(3): 2511-2522, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30218457

RESUMO

Sodium selenite (SSE), a source of inorganic selenium, has been widely used as a clinical cancer treatment, but the precise molecular mechanisms of SSE remain to be elucidated. Our in vitro experiments have confirmed that SSE treatment causes a transient increase in intracellular reactive oxygen species (ROS) levels, resulting in the inhibition of nuclear transcription factor-κB (NF-κB) signaling and p65 and nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha phosphorylation levels in 4T1 cells. The inhibition of NF-κB subsequently increased the expression of the apoptosis gene B-cell lymphoma-2-associated X (Bax) and downregulated the transcription of antiapoptosis genes, such as B-cell lymphoma-2, cellular inhibitor of apoptosis 1, and X-linked inhibitor of apoptosis. Additionally, the accumulation of ROS caused mitochondrial dysfunction, leading to the activation of caspase-9 and -3, thereby resulting in apoptosis. However, modulation of the ROS level by the chemical inhibitor N-acetyl-cysteine reversed these events. Similarly, in vitro murine syngeneic breast tumor models showed that SSE inhibits tumor growth by promoting apoptosis. These results indicate that SSE induces apoptosis via ROS-mediated inhibition of NF-κB signaling and activation of the Bax-caspase-9-caspase-3 axis.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Neoplasias Mamárias Animais/tratamento farmacológico , Selenito de Sódio/farmacologia , Animais , Apoptose/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Caspase 3/genética , Caspase 9/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/patologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , NF-kappa B/genética , Fosforilação/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA/genética , Proteína X Associada a bcl-2/genética
14.
Inflammation ; 42(2): 650-657, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30406463

RESUMO

Endometritis is a common inflammatory disease which endangers human and animal reproductive health. MicroRNA (miRNA) let-7c plays an important role in the inflammatory process; however, the regulatory underlying mechanism of let-7c in endometritis is unclear. In this study, we confirmed that let-7c was significantly reduced in LPS-induced mouse endometritis model, and overexpression of let-7c was able to effectively reduce uterine tissue damage caused by lipopolysaccharide (LPS), and then, a LPS-induced bovine endometrial epithelial cell (BEND) line was used to mimic the inflammatory model in vitro. Our data showed that overexpression of let-7c significantly reduced the expression of pro-inflammatory cytokines in BEND cells induced by LPS. Meanwhile, immunofluorescence and western blotting results showed that let-7c significantly inhibited LPS-induced phosphorylation of NF-κB, thereby inhibiting downstream pro-inflammatory cytokine expression. Taken together, our results suggested that let-7c ameliorates LPS-induced endometritis by attenuating the expression of pro-inflammatory cytokines via inhibition of the activation of NF-κB.


Assuntos
Endometrite/prevenção & controle , MicroRNAs/farmacologia , NF-kappa B/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Animais , Bovinos , Citocinas/metabolismo , Células Epiteliais/metabolismo , Feminino , Humanos , Lipopolissacarídeos/efeitos adversos , Camundongos , NF-kappa B/metabolismo , Fosforilação
15.
Biochem Biophys Res Commun ; 505(1): 1-6, 2018 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-30224056

RESUMO

Previous studies have shown that shikonin(SHI), the bioactive naphthoquinone constituent extracted from Chinese herb Lithospermum Erythrorhizon, possesses the potential to confront inflammation, and has little concerns towards drug residues comparing with antibiotics. While mastitis in dairy industry always trigger great harm to milk yields, effects of SHI on lipopolysaccharides (LPS)-induced mastitis should be measured. Here, we demonstrate anti-inflammatory effects of SHI on LPS-challenged mastitis and elucidate the potential signaling pathway both in vivo and in vitro. As a result, SHI administration mice significantly suffered less impairment of mammary gland and less recruitment of neutrophils than LPS administration mice. SHI significantly suppressed the expression of p-IκBα and p-p65, which are the critical proteins functioning in NF-kB signaling pathway. qPCR results indicate decreasing level of upstream pro-inflammatory cytokines in tissues, such as TNF-α, IL-1ß, and IL-6. The results are corresponding with the results in vitro, suggesting the potential usage of SHI as a therapeutic medicine in mastitis.


Assuntos
Mastite/prevenção & controle , NF-kappa B/metabolismo , Naftoquinonas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Linhagem Celular Tumoral , Citocinas/genética , Citocinas/metabolismo , Feminino , Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Mastite/induzido quimicamente , Mastite/metabolismo , Camundongos , Estrutura Molecular , Naftoquinonas/química , Peroxidase/metabolismo
16.
Int Immunopharmacol ; 64: 140-150, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30173054

RESUMO

Barbaloin is the major anthraquinone compound that is isolated from the leaf exudates of Aloe vera and is often used as a bittering agent in alcoholic beverages. Here, we investigated the potential protective role of barbaloin in a mouse model of lipopolysaccharide (LPS)-induced acute lung injury (ALI) and clarified the underlying mechanism in vitro. Histological analysis showed that barbaloin exhibited a certain protective effect on LPS-induced ALI. To further elucidate the mechanisms underlying the actions of barbaloin, LPS-stimulated macrophages were used in this study. The results showed that barbaloin decreased the phosphorylation levels of IκBα and NF-κB p65, leading to a reduction in the expression of pro-inflammatory cytokines (TNF-α, IL-1ß and IL-6). Furthermore, barbaloin also reduced the levels of intracellular reactive oxygen species (ROS) similarly to the antioxidant N­acetyl­l­cysteine (NAC), which alone repressed the LPS-induced phosphorylation of phosphoinositide 3-kinase (PI3K) and AKT. Additionally, a pharmacological inhibitor of PI3K/AKT, LY294002, also restrained the phosphorylation levels of IκBα and NF-κB p65 and thereby decreased the expression of pro-inflammatory cytokines. Together, these results show that barbaloin possesses a protective effect on LPS-induced ALI via suppressing the release of pro-inflammatory cytokines through the ROS-mediated PI3K/AKT/NF-κB pathway.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Antracenos/uso terapêutico , Lipopolissacarídeos/toxicidade , NF-kappa B/antagonistas & inibidores , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Lesão Pulmonar Aguda/induzido quimicamente , Animais , Antracenos/farmacologia , Anti-Inflamatórios/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptor 4 Toll-Like/fisiologia
17.
Front Immunol ; 9: 1916, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30186287

RESUMO

Bovine endometritis affects milk production and reproductive performance in dairy cows and causes serious economic loss. The underlying molecular mechanisms or signaling pathways of bovine endometritis remain unclear. In this study, we attempted to determine the expression mechanism of mir-223 in endometritis of dairy cows and evaluate its potential therapeutic value. We first confirmed that there was an increased level of miR-223 in endometritis, and then, an LPS-induced bovine endometrial epithelial cell (BEND) line was used to mimic the inflammatory model in vitro. Our data showed that activation of NF-κB promoted the transcription of miR-223, thus inhibiting activation of the inflammatory mediator NLRP3 and its mediation of IL-1ß production to protect against inflammatory damage. Meanwhile, in vivo studies showed that inhibition of mir-223 resulted in an enhanced pathology of mice during LPS-induced endometritis, while overexpression of mir-223 attenuated the inflammatory conditions in the uterus. In summary, our study highlights that miR-223 serves both to constrain the level of NLRP3 activation and to act as a protective factor in the inflammatory response and thus provides a future novel therapeutic modality for active flares in cow endometritis and other inflammatory diseases.


Assuntos
Doenças dos Bovinos/imunologia , Endometrite/imunologia , Endométrio/imunologia , Inflamassomos/imunologia , MicroRNAs/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Animais , Bovinos , Doenças dos Bovinos/patologia , Doenças dos Bovinos/terapia , Endometrite/patologia , Endometrite/terapia , Endometrite/veterinária , Endométrio/patologia , Feminino , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Transdução de Sinais/imunologia
18.
Inflamm Res ; 67(11-12): 903-911, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30145653

RESUMO

BACKGROUND: Nuciferine, a major bioactive component from the lotus leaf, has been reported to have notable anti-inflammatory activities such as renal inflammation and acute lung injury in previous studies. Mastitis is one of the most prevalent diseases in the dairy cattle, which causes large economic losses for the dairy industry. However, the effects of nuciferine on lipopolysaccharide (LPS)-induced mastitis have not been reported. METHODS AND RESULTS: Here, we investigated the anti-inflammatory effects of nuciferine on LPS-induced mastitis in mice and illuminated its potential mechanism on the TLR4-mediated signaling pathway in mouse mammary epithelial cells (mMECs). Histopathological changes and myeloperoxidase (MPO) activity assay showed that nuciferine treatment significantly alleviated the LPS-induced injury of mammary gland flocculus, inflammatory cells infiltration. qPCR and ELISA assays indicated that nuciferine dose-dependently reduced the levels of TNF-α and IL-1ß, which indicated that nuciferine might have therapeutic effects on mastitis. Furthermore, nuciferine treatment significantly decreased the expression of TLR4 in a dose-dependent manner. Besides, nuciferine was also found to suppress LPS-induced NF-κB activation. CONCLUSION: These findings indicate that nuciferine potently ameliorates LPS-induced mastitis by inhibition of the TLR4-NF-κB signaling pathway.


Assuntos
Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Aporfinas/farmacologia , Aporfinas/uso terapêutico , Mastite/tratamento farmacológico , NF-kappa B/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lipopolissacarídeos , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Mastite/induzido quimicamente , Mastite/metabolismo , Mastite/patologia , Camundongos Endogâmicos BALB C , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
19.
Int Immunopharmacol ; 61: 156-161, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29879659

RESUMO

Endometritis is the inflammation of the endometrium that is associated with lower conception rates, increased intervals from calving to first service, and more culls for failure to conceive, which leads to serious economic losses in the dairy industry. Leonurine, a natural active compound of Leonurus cardiaca, has been proved to possess various biological activities. However, there is still no study about its anti-inflammatory effects on LPS-induced endometritis. The present study aimed to demonstrate the underlying mechanism responsible for the anti-inflammatory effects of leonurine on LPS induced endometritis in mice and in bovine endometrial epithelial cells (bEECs). The results of pathological section displayed that leonurine alleviated LPS induced uterine injury. qRT-PCR and ELISA experiments suggested that leonurine inhibited the expression levels of TNF-α and IL-1ß in uterus tissues and bEECs. Molecular studies showed that TLR4 expression and nuclear factor (NF)-κB activation were both inhibited by leonurine treatment. These results suggested that the therapeutic effects of leonurine on LPS-induced endometritis in mice and bEECs may act by inhibiting the expression of TLR4 and its downstream mediated NF-κB pathway. Accordingly, leonurine may serve as an effective drug in preventing and treating LPS induced endometritis.


Assuntos
Anti-Inflamatórios/uso terapêutico , Endometrite/tratamento farmacológico , Endométrio/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Ácido Gálico/análogos & derivados , Animais , Bovinos , Células Cultivadas , Modelos Animais de Doenças , Endometrite/induzido quimicamente , Endométrio/patologia , Células Epiteliais/patologia , Feminino , Ácido Gálico/uso terapêutico , Interleucina-1beta/metabolismo , Leonurus/imunologia , Lipopolissacarídeos/imunologia , Camundongos , NF-kappa B/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
20.
Int J Biol Sci ; 14(6): 622-632, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29904277

RESUMO

Breast cancer is one of the most lethal cancers in the world. The fight against breast cancer has also become a major task for medical workers. MicroRNAs (miRNAs) are often aberrantly expressed in diverse cancers and are involved in progression and metastasis. Many studies have found that miRNAs can act as oncogenes or as tumor suppressor genes. Here, we show that miR-433 is significantly decreased in breast cancer cells. In addition, we demonstrate the effects of miR-433 on breast cancer cell apoptosis, migration and proliferation in an attempt to elucidate the mechanism of action of miR-433. Moreover, Rap1a was predicted to be a potential target of miR-433 using bioinformatic approaches, and we found that the expression of Rap1a is inversely correlated with the level of miR-433. Further studies through overexpression and knockdown of Rap1a confirmed that Rap1a, as a direct target gene of miR-433, contributes to the functions of miR-433. In addition, we found that Rap1a activates the MAPK signaling pathway, which can contribute to cell migration and proliferation and can inhibit apoptosis. Overall, these findings highlight miR-433 as a tumor suppressor gene in the regulation of the progression and metastatic potential of breast cancer and may benefit the future development of therapies targeting miR-433 in breast cancer.


Assuntos
Apoptose/fisiologia , Neoplasias da Mama/metabolismo , MicroRNAs/metabolismo , Animais , Apoptose/genética , Western Blotting , Neoplasias da Mama/genética , Movimento Celular/genética , Movimento Celular/fisiologia , Proliferação de Células/genética , Proliferação de Células/fisiologia , Feminino , Humanos , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/genética , Proteínas rap1 de Ligação ao GTP/genética , Proteínas rap1 de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA