The impact of chronic blackberry intake on the neuroinflammatory status of rats fed a standard or high-fat diet

2015 ◽  
Vol 26 (11) ◽  
pp. 1166-1173 ◽  
Author(s):  
Manuela Meireles ◽  
Cláudia Marques ◽  
Sónia Norberto ◽  
Iva Fernandes ◽  
Nuno Mateus ◽  
...  
Keyword(s):  
High Fat ◽  
2021 ◽  
Author(s):  
Qiao Jie ◽  
Yue-Zhong Ren ◽  
Yi-wen Wu

High-fat diets(HFD)are defined as lipids accounting for exceeded 30% of total energy in-take, and current research is mostly 45% and 60%. With a view of the tendency that patients who...


2005 ◽  
Vol 332 (1) ◽  
pp. 142-148 ◽  
Author(s):  
Anthony G. Passerini ◽  
Congzhu Shi ◽  
Nadeene M. Francesco ◽  
Peiying Chuan ◽  
Elisabetta Manduchi ◽  
...  

2018 ◽  
Vol 19 (3) ◽  
pp. S10
Author(s):  
B. Menta ◽  
A. Nicol ◽  
J. Ryals ◽  
M. Winter ◽  
K. McCarson ◽  
...  

2017 ◽  
Vol 29 (10) ◽  
pp. e12528 ◽  
Author(s):  
M. van den Top ◽  
F.-Y. Zhao ◽  
R. Viriyapong ◽  
N. J. Michael ◽  
A. C. Munder ◽  
...  

2019 ◽  
Vol 74 ◽  
pp. 121-134 ◽  
Author(s):  
Sarah J. Spencer ◽  
Bashirah Basri ◽  
Luba Sominsky ◽  
Alita Soch ◽  
Monica T. Ayala ◽  
...  

2019 ◽  
Vol 317 (2) ◽  
pp. E298-E311 ◽  
Author(s):  
Colin S. McCoin ◽  
Alex Von Schulze ◽  
Julie Allen ◽  
Kelly N. Z. Fuller ◽  
Qing Xia ◽  
...  

The impact of sexual dimorphism and mitophagy on hepatic mitochondrial adaptations during the treatment of steatosis with physical activity are largely unknown. Here, we tested if deficiencies in liver-specific peroxisome proliferative activated-receptor-γ coactivator-1α (PGC-1α), a transcriptional coactivator of biogenesis, and BCL-2/ADENOVIRUS EIB 19-kDa interacting protein (BNIP3), a mitophagy regulator, would impact hepatic mitochondrial adaptations (respiratory capacity, H2O2production, mitophagy) to a high-fat diet (HFD) and HFD plus physical activity via voluntary wheel running (VWR) in both sexes. Male and female wild-type (WT), liver-specific PGC-1α heterozygote (LPGC-1α), and BNIP3 null mice were thermoneutral housed (29–31°C) and divided into three groups: sedentary-low-fat diet (LFD), 16 wk of (HFD), or 16 wk of HFD with VWR for the final 8 wk (HFD + VWR) ( n = 5–7/sex/group). HFD did not impair mitochondrial respiratory capacity or coupling in any group; however, HFD + VWR significantly increased maximal respiratory capacity only in WT and PGC-1α females. Males required VWR to elicit mitochondrial adaptations that were inherently present in sedentary females including greater mitochondrial coupling control and reduced H2O2production. Females had overall reduced markers of mitophagy, steatosis, and liver damage. Steatosis and markers of liver injury were present in sedentary male mice on the HFD and were effectively reduced with VWR despite no resolution of steatosis. Overall, reductions in PGC-1α and loss of BNIP3 only modestly impacted mitochondrial adaptations to HFD and HFD + VWR with the biggest effect seen in BNIP3 females. In conclusion, hepatic mitochondrial adaptations to HFD and treatment of HFD-induced steatosis with VWR are more dependent on sex than PGC-1α or BNIP3.


Toxicology ◽  
2019 ◽  
Vol 414 ◽  
pp. 27-34 ◽  
Author(s):  
Weihong Xu ◽  
Jie Yu ◽  
Zhigang Jiang ◽  
Wenxia Yan ◽  
Shengnan Li ◽  
...  

Hepatology ◽  
2009 ◽  
Vol 51 (6) ◽  
pp. 2234-2235 ◽  
Author(s):  
Adriana L. Burgueño ◽  
Julieta Carabelli ◽  
Silvia Sookoian ◽  
Carlos J. Pirola

2018 ◽  
Vol 115 (38) ◽  
pp. E8996-E9005 ◽  
Author(s):  
Brandon M. Gassaway ◽  
Max C. Petersen ◽  
Yulia V. Surovtseva ◽  
Karl W. Barber ◽  
Joshua B. Sheetz ◽  
...  

Insulin resistance drives the development of type 2 diabetes (T2D). In liver, diacylglycerol (DAG) is a key mediator of lipid-induced insulin resistance. DAG activates protein kinase C ε (PKCε), which phosphorylates and inhibits the insulin receptor. In rats, a 3-day high-fat diet produces hepatic insulin resistance through this mechanism, and knockdown of hepatic PKCε protects against high-fat diet-induced hepatic insulin resistance. Here, we employed a systems-level approach to uncover additional signaling pathways involved in high-fat diet-induced hepatic insulin resistance. We used quantitative phosphoproteomics to map global in vivo changes in hepatic protein phosphorylation in chow-fed, high-fat–fed, and high-fat–fed with PKCε knockdown rats to distinguish the impact of lipid- and PKCε-induced protein phosphorylation. This was followed by a functional siRNA-based screen to determine which dynamically regulated phosphoproteins may be involved in canonical insulin signaling. Direct PKCε substrates were identified by motif analysis of phosphoproteomics data and validated using a large-scale in vitro kinase assay. These substrates included the p70S6K substrates RPS6 and IRS1, which suggested cross talk between PKCε and p70S6K in high-fat diet-induced hepatic insulin resistance. These results identify an expanded set of proteins through which PKCε may drive high-fat diet-induced hepatic insulin resistance that may direct new therapeutic approaches for T2D.


Sign in / Sign up

Export Citation Format

Share Document