lipid secretion
Recently Published Documents


TOTAL DOCUMENTS

252
(FIVE YEARS 8)

H-INDEX

33
(FIVE YEARS 3)

2021 ◽  
Vol 36 ◽  
pp. 102537
Author(s):  
Jiayi Yang ◽  
Lei Shi ◽  
Detian Xu ◽  
Jia Liu ◽  
Linglin Zhang ◽  
...  

2020 ◽  
Author(s):  
Lingling Zhang ◽  
Jiayi Yang ◽  
Xiaojing Liu ◽  
Detian Xu ◽  
Lei Shi ◽  
...  

Abstract Background: Acne vulgaris is a chronic inflammatory cutaneous disease. 5-Aminolaevulinic acid photodynamic therapy (ALA-PDT) is a novel and effective therapy for severe acne vulgaris. However, the specific mechanism of ALA-PDT for acne still remain unclear. Here, we investigate the possible mechanism of intense inflammatory response of ALA-PDT for acne vulgaris. Results: It appeared that ALA-PDT suppresses proliferation and lipid secretion of primary human sebocytes. And ALA-PDT could up-regulate the expression of CXCL8 in vivo and in vitro, amplifying inflammatory response by recruiting T cells, B cells, neutrophils and macrophages. We also found that ALA-PDT elevated the expression of CXCL8 via p38 pathway. SB203580, a p38 pathway inhibitor, decreased the expression of CXCL8 after ALA-PDT in sebocytes. Conclusion: ALA-PDT amplifies intense inflammatory response in the treatment of acne vulgaris via CXCL8. Our data deciphers the mechanism of intense inflammatory response after ALA-PDT for acne vulgaris.


2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Limin Zhao ◽  
Hao Ke ◽  
Haibo Xu ◽  
Guo-Dong Wang ◽  
Honglei Zhang ◽  
...  

2019 ◽  
Vol 70 (1) ◽  
pp. e812-e813
Author(s):  
Friederike Poppenborg ◽  
Caroline Bopp ◽  
Annika Nicklas ◽  
Frank Lammert ◽  
Susanne N Weber

2019 ◽  
Vol 5 (1) ◽  
pp. eaau3795 ◽  
Author(s):  
Da-Wei Li ◽  
Srinivasan Balamurugan ◽  
Yu-Feng Yang ◽  
Jian-Wei Zheng ◽  
Dan Huang ◽  
...  

Commercialization of algal lipids and biofuels is still impractical owing to the unavailability of lipogenic strains and lack of economically viable oil extraction strategies. Because lipogenesis is governed by multiple factors, success in generating industrial-suitable algal strains using conventional strategies has been limited. We report the discovery of a novel bZIP1 transcription factor, NobZIP1, whose overexpression results in a remarkable elevation of lipid accumulation and lipid secretion in a model microalgaNannochloropsis oceanica, without impairing other physiological properties. Chromatin immunoprecipitation–quantitative PCR analysis revealed that the key genes up- and down-regulated by NobZIP1 are involved in lipogenesis and cell wall polymer synthesis, respectively, which, in turn, induce lipid overproduction and secretion. Among these regulated genes, UDP-glucose dehydrogenase was shown to alter cell wall composition, thus also boosting lipid secretion. In summary, these results offer a comprehensive strategy for concurrent lipid overproduction and secretion, strongly increasing the commercial potential of microalgae.


mBio ◽  
2018 ◽  
Vol 9 (5) ◽  
Author(s):  
Asmaa Tazi ◽  
João Ricardo Araujo ◽  
Céline Mulet ◽  
Ellen T. Arena ◽  
Giulia Nigro ◽  
...  

ABSTRACT The gut microbiota contributes to nutrients absorption and metabolism by enterocytes, but the molecular mechanisms involved remain poorly understood, and most conclusions are inferred from studies comparing germfree and conventional animals colonized with diverse bacterial species. We selected two model commensal microorganisms, Escherichia coli and Lactobacillus paracasei, to assess the role of the small-intestinal microbiota in modulating lipid absorption and metabolism by the epithelium. Using an integrated approach encompassing cellular and murine models and combining metabolic parameters measurement, lipid droplet imaging, and gene expression analysis, we demonstrated that under homeostatic conditions, L. paracasei promotes fat storage in enterocytes, whereas E. coli enhances lipid catabolism and reduces chylomicron circulating levels. The Akt/mammalian target of sirolimus (mTOR) pathway is inhibited by both bacterial species in vitro, indicating that several regulatory pathways are involved in the distinct intracellular lipid outcomes associated with each bacterial species. Moreover, soluble bacterial factors partially reproduce the effects observed with live microorganisms. However, reduction of chylomicron circulating levels in E. coli-colonized animals is lost under high-fat-diet conditions, whereas it is potentiated by L. paracasei colonization accompanied by resistance to hypercholesterolemia and excess body weight gain. IMPORTANCE The specific contribution of each bacterial species within a complex microbiota to the regulation of host lipid metabolism remains largely unknown. Using two model commensal microorganisms, L. paracasei and E. coli, we demonstrated that both bacterial species impacted host lipid metabolism in a diet-dependent manner and, notably, that L. paracasei-colonized mice but not E. coli-colonized mice resisted high-fat-diet-induced body weight gain. In addition, we set up cellular models of fatty acid absorption and secretion by enterocytes cocultured with bacteria and showed that, in vitro, both L. paracasei and E. coli inhibited lipid secretion, through increased intracellular fat storage and enhanced lipid catabolism, respectively.


2018 ◽  
Vol 68 ◽  
pp. S7-S8
Author(s):  
R. Charlotte ◽  
B. Annika ◽  
M. Krawczyk ◽  
F. Lammert ◽  
S.N. Weber

Sign in / Sign up

Export Citation Format

Share Document