Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Anim Biotechnol ; 34(8): 3808-3818, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37435839

RESUMO

In the dairy industry, glucose (Glu) is used as bioactive substance to increase milk yield. However, the molecular regulation underneath needs further clarification. Here, the regulation and its molecular mechanism of Glu on cell growth and casein synthesis of dairy cow mammary epithelial cells (DCMECs) were investigated. When Glu was added from DCMECs, both cell growth, ß-casein expression and the mechanistic target of rapamycin complex 1 (mTORC1) pathway were increased. Overexpression and silencing of mTOR revealed that Glu promoted cell growth and ß-casein expression through the mTORC1 pathway. When Glu was added from DCMECs, both Adenosine 5'-monophosphate-activated protein kinase α (AMPKα) and Sestrin2 (SESN2) expression were decreased. Overexpression and silencing of AMPKα or SESN2 uncovered that AMPKα suppressed cell growth and ß-casein synthesis through inhibiting mTORC1 pathway, and SESN2 suppressed cell growth and ß-casein synthesis through activating AMPK pathway. When Glu was depleted from DCMECs, both activating transcription factor 4 (ATF4) and nuclear factor (erythroid-derived 2)-like 2 (Nrf2) expression were increased. Overexpression or silencing of ATF4 or Nrf2 demonstrated that Glu depletion promoted SESN2 expression through ATF4 and Nrf2. Together, these results indicate that in DCMECs, Glu promoted cell growth and casein synthesis via ATF4/Nrf2-SESN2-AMPK-mTORC1 pathway.


Assuntos
Fator 4 Ativador da Transcrição , Caseínas , Feminino , Bovinos , Animais , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Fator 4 Ativador da Transcrição/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Glucose/farmacologia , Glucose/metabolismo , Glândulas Mamárias Animais/metabolismo , Células Epiteliais/metabolismo
2.
Ecotoxicol Environ Saf ; 214: 112078, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33676053

RESUMO

It is well known that the dairy cow production is very sensitive to environmental factors, including high temperature, high humidity and radiant heat sources. High temperature-induced heat stress is the main environmental factor that causes oxidative stress and apoptosis, which affects the development of mammary glands in dairy cows. Dihydromyricetin (DMY) is a nature flavonoid compound extracted from Ampelopsis grossedentata; it has been shown to have various pharmacological functions, such as anti-inflammation, antitumor and liver protection. The present study aims to evaluate the protective effect of DMY on heat stress-induced dairy cow mammary epithelial cells (DCMECs) apoptosis and explore the potential mechanisms. The results show that heat stress triggers heat shock response and reduces cell viability in DCMECs; pretreatment of DCMECs with DMY (25 µM) for 12 h significantly alleviates the negative effects of heat stress on cells. DMY can provide cytoprotective effects by suppressing heat stress-caused mitochondrial membrane depolarization and mitochondrial dysfunction, Bax and Caspase 3 activity, and modulation of oxidative enzymes, thereby preventing ROS production and apoptosis in DCMECs. Importantly, DMY treatment could attenuate heat stress-induced mitochondrial fragmentation through mediating the expression of mitochondrial fission and fusion-related genes, including Dynamin related protein 1 (Drp1), Mitochondrial fission 1 protein (Fis1), and Mitofusin1, 2 (Mfn1, 2). Above all, our findings demonstrate that DMY could protect DCMECs against heat stress-induced injury through preventing oxidative stress, the imbalance of mitochondrial fission and fusion, which provides useful evidence that DMY can be a promising therapeutic drug for protecting heat stress-induced mammary glands injury and mastitis.


Assuntos
Flavonóis/farmacologia , Resposta ao Choque Térmico/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Animais , Apoptose/efeitos dos fármacos , Bovinos , Sobrevivência Celular/efeitos dos fármacos , Dinaminas , Células Epiteliais/efeitos dos fármacos , Feminino , Mitocôndrias/efeitos dos fármacos , Dinâmica Mitocondrial/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos
3.
J Dairy Res ; 86(2): 181-187, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31122298

RESUMO

This research paper addresses the hypothesis that Septin6 is a key regulatory factor influencing amino acid (AA)-mediated cell growth and casein synthesis in dairy cow mammary epithelial cells (DCMECs). DCMECs were treated with absence of AA (AA-), restricted concentrations of AA (AAr) or normal concentrations of AA (AA+) for 24 h. Cell growth, expression of CSN2 and Septin6 were increased in response to AA supply. Overexpressing or inhibiting Septin6 demonstrated that cell growth, expression of CSN2, mTOR, p-mTOR, S6K1 and p-S6K1 were up-regulated by Septin6. Furthermore, overexpressing or inhibiting mTOR demonstrated that the increase in cell growth and expression of CSN2 in response to Septin6 overexpression were inhibited by mTOR inhibition, and vice versa. Our hypothesis was supported; we were able to show that Septin6 is an important positive factor for cell growth and casein synthesis, it up-regulates AA-mediated cell growth and casein synthesis through activating mTORC1 pathway in DCMECs.


Assuntos
Aminoácidos/farmacologia , Caseínas/metabolismo , Células Epiteliais/metabolismo , Glândulas Mamárias Animais/citologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Septinas/metabolismo , Animais , Caseínas/genética , Bovinos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Septinas/genética
4.
J Dairy Res ; 85(4): 439-444, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30088470

RESUMO

Sterol regulatory element binding protein 1 (SREBP1) has a central regulatory effect on milk fat synthesis. Lipopolysaccharides (LPS) can induce mastitis and cause milk fat depression in cows. SREBP1 is also known to be associated with inflammatory regulation. Thus, in the current study, we hypothesized that LPS-induced milk fat depression in dairy cow mammary epithelial cells (DCMECs) operates via decreased SREBP1 expression and activity. To examine the hypothesis, DCMECs were isolated and purified from dairy cow mammary tissue and treated with LPS (10 µg/ml). LPS treatment of DCMECs suppressed lipid-metabolism-related transcription factor SREBP1 mRNA expression, nuclear translocation and protein expression, leading to reduced triglyceride content. The transcription levels of acetyl-CoA carboxylase-1 and fatty acid synthetase were significantly down-regulated in DCMECs after LPS treatment, suggesting that acetyl-CoA carboxylase-1 and fatty acid synthetase involved in de novo milk fat synthesis was regulated by SREBP1. In summary, these results suggest that LPS induces milk fat depression in dairy cow mammary epithelial cells via decreased expression of SREBP1 in a time-dependent manner.


Assuntos
Bovinos/fisiologia , Células Epiteliais/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Glândulas Mamárias Animais/citologia , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Triglicerídeos/metabolismo , Animais , Células Epiteliais/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Lipogênese/efeitos dos fármacos , Proteína de Ligação a Elemento Regulador de Esterol 1/genética
5.
Int J Mol Sci ; 16(7): 16622-41, 2015 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-26204835

RESUMO

As a protective factor for lipopolysaccharide (LPS)-induced injury, 14-3-3γ has been the subject of recent research. Nevertheless, whether 14-3-3γ can regulate lactation in dairy cow mammary epithelial cells (DCMECs) induced by LPS remains unknown. Here, the anti-inflammatory effect and lactation regulating ability of 14-3-3γ in LPS-induced DCMECs are investigated for the first time, and the molecular mechanisms responsible for their effects are explored. The results of qRT-PCR showed that 14-3-3γ overexpression significantly inhibited the mRNA expression of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), interleukin-1ß (IL-1ß) and inducible nitric oxide synthase (iNOS). Enzyme-linked immunosorbent assay (ELISA) analysis revealed that 14-3-3γ overexpression also suppressed the production of TNF-α and IL-6 in cell culture supernatants. Meanwhile, CASY-TT Analyser System showed that 14-3-3γ overexpression clearly increased the viability and proliferation of cells. The results of kit methods and western blot analysis showed that 14-3-3γ overexpression promoted the secretion of triglycerides and lactose and the synthesis of ß-casein. Furthermore, the expression of genes relevant to nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPKs) and lactation-associated proteins were assessed by western blot, and the results suggested that 14-3-3γ overexpression inactivated the NF-κB and MAPK signaling pathways by down-regulating extracellular signal regulated protein kinase (ERK), p38 mitogen-activated protein kinase (p38MAPK) and inhibitor of NF-κB (IκB) phosphorylation levels, as well as by inhibiting NF-κB translocation. Meanwhile, 14-3-3γ overexpression enhanced the expression levels of ß-casein, mammalian target of rapamycin (mTOR), ribosomal protein S6 kinase 1 (S6K1), serine/threonine protein kinase Akt 1 (AKT1), sterol regulatory element binding protein 1 (SREBP1) and peroxisome proliferator-activated receptor gamma (PPARγ). These results suggest that 14-3-3γ was able to attenuate the LPS-induced inflammatory responses and promote proliferation and lactation in LPS-induced DCMECs by inhibiting the activation of the NF-κB and MAPK signaling pathways and up-regulating mTOR signaling pathways to protect against LPS-induced injury.


Assuntos
Proteínas 14-3-3/metabolismo , Caseínas/metabolismo , Células Epiteliais/metabolismo , Sistema de Sinalização das MAP Quinases , Glândulas Mamárias Animais/metabolismo , NF-kappa B/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas 14-3-3/genética , Animais , Caseínas/genética , Bovinos , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Interleucinas/genética , Interleucinas/metabolismo , Lactose/metabolismo , Lipopolissacarídeos/farmacologia , Glândulas Mamárias Animais/citologia , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
In Vitro Cell Dev Biol Anim ; 56(4): 322-331, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32377999

RESUMO

Heat stress-induced reductions in milk yield and the dysfunction of mammary glands are economically important challenges that face the dairy industry, especially during summer. The aim of the present study is to investigate the effects of heat stress on mitochondrial function by using dairy cow mammary epithelial cells (DCMECs) as an in vitro model. Live cell imaging shows that the mitochondria continually change shape through fission and fusion. However, heat stress induces the fragmentation of mitochondria, as well as the decreased of ATP level, membrane potential, and anti-oxidant enzyme activity and the increased of respiratory chain complex I activity. In addition, the cytosolic Ca2+ concentration and cytochrome c expression (Cyto-c) were increased after heat stress treatment. Both qRT-PCR and western blot analysis indicate that mitofusin1/2 (Mfn1/2) and optic atrophy protein-1 (Opa-1) are downregulated after heat stress, whereas dynamin-related protein 1 (Drp1) and fission 1 (Fis-1) are upregulated, which explains the observed defect of mitochondrial network dynamics. Accordingly, the present study indicated that heat stress induced the dysfunction of DCMEC through disruption of the normal balance of mitochondrial fission and fusion.


Assuntos
Apoptose , Indústria de Laticínios , Células Epiteliais/patologia , Resposta ao Choque Térmico , Glândulas Mamárias Animais/patologia , Mitocôndrias/metabolismo , Animais , Cálcio/metabolismo , Bovinos , Citocromos c/metabolismo , Transporte de Elétrons , Células Epiteliais/metabolismo , Feminino , Potencial da Membrana Mitocondrial , Dinâmica Mitocondrial , Estresse Oxidativo
7.
J Agric Food Chem ; 66(45): 12000-12008, 2018 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-30375228

RESUMO

Cell proliferation and casein synthesis of dairy cow mammary epithelial cells (DCMECs) are regulated by many factors. This research aimed to investigate the effect of 14-3-3ε on cell proliferation and casein synthesis in DCMECs and to reveal the underlying mechanism. Overexpressing or inhibiting 14-3-3ε demonstrated that cell proliferation; casein synthesis; expression of mTOR, p-mTOR, S6K1, and p-S6K1; and lysosomal localization of mTOR were all up-regulated by 14-3-3ε overexpressing and down-regulated by 14-3-3ε inhibiting. In addition, inhibiting mTOR demonstrated that the up-regulation of cell proliferation and casein synthesis in response to 14-3-3ε overexpressing was removed by inhibiting mTOR. Furthermore, the regulatory mechanism of 14-3-3ε was analyzed by coimmunoprecipitation, and we found that 14-3-3ε could interact with PI3K and activate mTORC1 pathway via PI3K. In addition, DCMECs were treated with insulin and prolactin, and the result showed that the cell proliferation and the expression of CSN2 and 14-3-3ε were all up-regulated by these hormones. In conclusion, the current research showed that 14-3-3ε is an important positive regulatory factor for cell proliferation and casein synthesis in DCMECs, as it up-regulates cell proliferation and casein synthesis via activating PI3K-mTOR pathway.


Assuntos
Proteínas 14-3-3/metabolismo , Caseínas/biossíntese , Bovinos/metabolismo , Células Epiteliais/metabolismo , Glândulas Mamárias Animais/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas 14-3-3/genética , Animais , Bovinos/genética , Proliferação de Células , Células Epiteliais/citologia , Feminino , Fosfatidilinositol 3-Quinases/genética , Transdução de Sinais , Serina-Treonina Quinases TOR/genética
8.
In Vitro Cell Dev Biol Anim ; 52(10): 1044-1059, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27287918

RESUMO

Peroxisome proliferator-activated receptor gamma (PPARγ) participates in lipogenesis in rats, goats, and humans. However, the exact mechanism of PPARγ regulation on milk fat synthesis in dairy cow mammary epithelial cells (DCMECs) remains largely unexplored. The aim of this study was to investigate the role of PPARγ regarding milk fat synthesis in DCMECs and to ascertain whether milk fat precursor acetic acid and palmitic acid could interact with PPARγ signaling to regulate milk fat synthesis. For this study, we examined the effects of PPARγ overexpression and gene silencing on cell growth, triacylglycerol synthesis, and the messenger RNA (mRNA) and protein expression levels of genes involved in milk fat synthesis in DCMECs. In addition, we investigated the influences of acetic acid and palmitic acid on the mRNA and protein levels of milk lipogenic genes and triacylglycerol synthesis in DCMECs transfected with PPARγ small interfering RNA (siRNA) and PPARγ expression vector. The results showed that when PPARγ was silenced, cell viability, proliferation, and triacylglycerol secretion were obviously reduced. Gene silencing of PPARγ significantly downregulated the expression levels of milk fat synthesis-related genes in DCMECs. PPARγ overexpression improved cell viability, proliferation, and triacylglycerol secretion. The expression levels of milk lipogenic genes were significantly increased when PPARγ was overexpressed. Acetic acid and palmitic acid could markedly improve triacylglycerol synthesis and upregulate the expression levels of PPARγ and other lipogenic genes in DCMECs. These results suggest that PPARγ is a positive regulator of milk fat synthesis in DCMECs and that acetic acid and palmitic acid could partly regulate milk fat synthesis in DCMECs via PPARγ signaling.


Assuntos
Indústria de Laticínios , Células Epiteliais/metabolismo , Lipídeos/biossíntese , Glândulas Mamárias Animais/citologia , Leite/química , PPAR gama/metabolismo , Ácido Acético/farmacologia , Animais , Bovinos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Inativação Gênica/efeitos dos fármacos , Lipogênese/efeitos dos fármacos , Lipogênese/genética , PPAR gama/genética , Ácido Palmítico/farmacologia , Triglicerídeos/metabolismo
9.
Exp Ther Med ; 11(4): 1279-1287, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27073437

RESUMO

Our previous study demonstrated that 14-3-3γ overexpression was able to inhibit the production of lipopolysaccharide (LPS)-induced cytokines in dairy cow mammary epithelial cells (DCMECs) by inhibiting the activation of nuclear factor-κB (NF-κB) signaling pathways. However, the association between 14-3-3γ overexpression and milk fat synthesis in LPS-induced DCMECs remains unclear. Therefore, the present study investigated the effect of 14-3-3γ on cell viability and milk fat synthesis in LPS-induced DCMECs. The results of the MTT assay and lactate dehydrogenase activity assay demonstrated that 14-3-3γ overexpression was able to attenuate LPS-induced cytotoxicity in DCMECs, and increase the viability of the cells. In addition, the results of reverse transcription-quantitative polymerase chain reaction suggested that mRNA expression levels of genes associated with milk fat synthesis, including sterol regulatory element binding protein (SREBP1), peroxisome proliferator-activated receptor-γ (PPARG), cluster of differentiation 36, acetyl-coA carboxylase (ACC), fatty acid synthase (FAS) and fatty acid binding protein-3, were significantly upregulated in cells overexpressing the 14-3-3γ protein. In addition, as compared with the LPS-treated group, the activities of FAS and ACC were significantly increased. Furthermore, western blotting demonstrated that 14-3-3γ overexpression enhanced the protein expression levels of phosphorylated SREBP1 and PPARG. These results suggested that high levels of 14-3-3γ protein were able to attenuate LPS-induced cell damage and promote milk fat synthesis in LPS-induced DCMECs by increasing the cell viability and upregulating the expression levels of transcription factors associated with milk fat synthesis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA