endothelial hyperpermeability
Recently Published Documents


TOTAL DOCUMENTS

89
(FIVE YEARS 24)

H-INDEX

17
(FIVE YEARS 6)

2021 ◽  
Vol 36 (1) ◽  
Author(s):  
Yu‐Hsuan Wu ◽  
Wei‐Chun Chen ◽  
Chin‐Kai Tseng ◽  
Yen‐Hsu Chen ◽  
Chun‐Kuang Lin ◽  
...  

2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Rui Zuo ◽  
Li-Feng Ye ◽  
Yi Huang ◽  
Zi-Qing Song ◽  
Lei Wang ◽  
...  

Abstract Background A recent study has reported that patients with nonalcoholic fatty liver disease (NAFLD) are more susceptible to coronary microvascular dysfunction (CMD), which may predict major adverse cardiac events. However, little is known regarding the causes of CMD during NAFLD. In this study, we aimed to explore the role of hepatic small extracellular vesicles (sEVs) in regulating the endothelial dysfunction of coronary microvessels during NAFLD. Results We established two murine NAFLD models by feeding mice a methionine-choline-deficient (MCD) diet for 4 weeks or a high-fat diet (HFD) for 16 weeks. We found that the NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome-dependent endothelial hyperpermeability occurred in coronary microvessels during both MCD diet and HFD-induced NAFLD. The in vivo and in vitro experiments proved that novel-microRNA(miR)-7-abundant hepatic sEVs were responsible for NLRP3 inflammasome-dependent endothelial barrier dysfunction. Mechanistically, novel-miR-7 directly targeted lysosomal associated membrane protein 1 (LAMP1) and promotes lysosomal membrane permeability (LMP), which in turn induced Cathepsin B-dependent NLRP3 inflammasome activation and microvascular endothelial hyperpermeability. Conversely, a specific novel-miR-7 inhibitor markedly improved endothelial barrier integrity. Finally, we proved that steatotic hepatocyte was a significant source of novel-miR-7-contained hepatic sEVs, and steatotic hepatocyte-derived sEVs were able to promote NLRP3 inflammasome-dependent microvascular endothelial hyperpermeability through novel-miR-7. Conclusions Hepatic sEVs contribute to endothelial hyperpermeability in coronary microvessels by delivering novel-miR-7 and targeting the LAMP1/Cathepsin B/NLRP3 inflammasome axis during NAFLD. Our study brings new insights into the liver-to-microvessel cross-talk and may provide a new diagnostic biomarker and treatment target for microvascular complications of NAFLD. Graphical Abstract


2021 ◽  
Author(s):  
Rui Zuo ◽  
Li-feng Ye ◽  
Yi Huang ◽  
Zi-qing Song ◽  
Lei Wang ◽  
...  

Abstract BackgroundA recent study has reported that patients with nonalcoholic fatty liver disease (NAFLD) are more susceptible to coronary microvascular dysfunction (CMD), which may predict major adverse cardiac events. However, little is known regarding the causes of CMD during NAFLD. In this study, we aimed to explore the role of hepatic small extracellular vesicles (sEVs) in regulating the endothelial dysfunction of coronary microvessels during NAFLD.ResultsWe established two murine NAFLD models by feeding mice a methionine-choline-deficient (MCD) diet for 4 weeks or a high-fat diet (HFD) for 16 weeks. We found that the NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome-dependent endothelial hyperpermeability occurred in coronary microvessels during both MCD diet and HFD-induced NAFLD. The in vivo and in vitro experiments proved that novel-miR-7-abundant hepatic sEVs were responsible for NLRP3 inflammasome-dependent endothelial barrier dysfunction. Mechanistically, novel-miR-7 directly targeted lysosomal associated membrane protein 1 (LAMP1) and promotes lysosomal membrane permeability (LMP), which in turn induced Cathepsin B-dependent NLRP3 inflammasome activation and microvascular endothelial hyperpermeability. Conversely, a specific novel-miR-7 inhibitor markedly improved endothelial barrier integrity. Finally, we proved that steatotic hepatocyte was a main resource of novel-miR-7-contained hepatic sEVs, and steatotic hepatocyte-derived sEVs were able to promote NLRP3 inflammasome-dependent microvascular endothelial hyperpermeability through novel-miR-7.ConclusionsHepatic sEVs contribute to endothelial hyperpermeability in coronary microvessels by delivering novel-miR-7 and targeting the LAMP1/Cathepsin B/NLRP3 inflammasome axis during NAFLD. Our study brings new insights into the liver-to-microvessel cross-talk and may provide a new diagnostic biomarker and treatment target for microvascular complications of NAFLD.


2021 ◽  
Vol Volume 14 ◽  
pp. 3321-3333
Author(s):  
Xiao Ran ◽  
Qin Zhang ◽  
Shaoping Li ◽  
Zhen Yu ◽  
Li Wan ◽  
...  

2021 ◽  
Author(s):  
Yan-nian Luo ◽  
Nan-nan He ◽  
Juan Xu ◽  
Rui Wang ◽  
Wen Cao ◽  
...  

Abstract The present study was aimed to explore the protective role of isoacteoside (ISO) in cecal ligation and puncture (CLP)-induced acute lung injury (ALI) in mice. Mice were divided into the following groups: sham control group, ALI group, and ALI+ISO group, in which mice received 10,50 or 100 mg/kg/day of ISO for 3 days before, 0h and 12h after CLP surgery. In the first experiment, all mice were maintained until 72 h after the CLP operation to calculate the survival rate. In the second experiment, mouse serum and lung and bronchoalveolar lavage fluid (BALF) were collected 24 h after model establishment for detection. The results revealed that ISO significantly improved the ALI associated survival rate, reduced the pathological injury, ALI score, infiltration of inflammatory cells, leakage of cells and proteins into BALF, systemic and local cytokine secretion, and pulmonary oxidative stress. Moreover, ISO significantly inhibited the expression levels of the pro-inflammatory proteins TLR4, MyD88, p-NF-κB p65, p-IKKαβ, and p-IκBα and increased the expression levels of the endothelial permeability related proteins ZO-1, claudin 5 and VE-cadherin. In conclusions, ISO mitigated acute lung injury in mice which was attributed to the capacity of ISO to inhibit inflammation, oxidative stress and endothelial hyperpermeability.


2021 ◽  
Vol 35 (3) ◽  
Author(s):  
Athanasios Ziogas ◽  
Md Sanaullah Sajib ◽  
Jong‐Hyung Lim ◽  
Tiago C. Alves ◽  
Anupam Das ◽  
...  

2020 ◽  
Vol 28 (23) ◽  
pp. 115809
Author(s):  
Vijaya Lakshmi Bodiga ◽  
Jagadeesh Bathula ◽  
Madhukar Rao Kudle ◽  
Praveen Kumar Vemuri ◽  
Sreedhar Bodiga

Author(s):  
Anoek L.I. van Leeuwen ◽  
David N. Naumann ◽  
Nicole A.M. Dekker ◽  
Peter L. Hordijk ◽  
Sam D. Hutchings ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document