Abstract

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-1β (IL-1β) 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 β-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 β-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.

Highlights

  • As a common metabolic disease, the incidence of sub-acute ruminal acidosis (SARA) is greater than20% in the primary and middle lactation of dairy cows, which causes significant economic damage to dairy cattle cultivation [1]

  • cytokeratin 18 (CK18) was counterstained with fluorescein isothiocyanate (FITC), and nuclei were counterstained with propidium iodide (PI)

  • Our results demonstrated that treatment of dairy cow mammary epithelial cells (DCMECs) with LPS alone resulted in a remarkable increase in tumor necrosis factor-α (TNF-α) and IL-6 production compared to control group (p < 0.01)

Read more

Summary

Introduction

As a common metabolic disease, the incidence of sub-acute ruminal acidosis (SARA) is greater than20% in the primary and middle lactation of dairy cows, which causes significant economic damage to dairy cattle cultivation [1]. LPS, as a major component of the outer membrane of Gram-negative bacteria, is usually viewed as a highly efficient proinflammatory response factor that activates the nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) pathways and results in the release of a large number of pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), IL-1β and so on [3,4,5,6]. Many studies have shown that proinflammatory cytokines in dairy cows can lead to chronic mastitis [7], changes in nutritional metabolism and cells apoptosis which can influence the milk composition and milk yield [8,9,10,11]. Several intracellular signaling-regulated molecules have been identified [12,13]

Objectives
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call