scholarly journals Design of a multi-sensor platform for integrating extracellular acidification rate with multi-metabolite flux measurement for small biological samples

2019 ◽  
Vol 133 ◽  
pp. 39-47 ◽  
Author(s):  
Yusra M. Obeidat ◽  
Ming-Hao Cheng ◽  
Giovana Catandi ◽  
Elaine Carnevale ◽  
Adam J. Chicco ◽  
...  
2021 ◽  
Vol 160 ◽  
pp. 105750
Author(s):  
Ramachandran Rajakumaran ◽  
Alagumalai Krishnapandi ◽  
Shen-Ming Chen ◽  
Karuppaiah Balamurugan ◽  
Fu Mao Chang ◽  
...  

Hypertension ◽  
2020 ◽  
Vol 76 (1) ◽  
pp. 121-132
Author(s):  
Xiao Chun Li ◽  
Xinchun Zhou ◽  
Jia Long Zhuo

The present study tested the hypotheses that overexpression of an intracellular Ang II (angiotensin II) fusion protein, mito-ECFP/Ang II, selectively in the mitochondria of mouse proximal tubule cells induces mitochondrial oxidative and glycolytic responses and elevates blood pressure via the Ang II/AT 1a receptor/superoxide/NHE3 (the Na + /H + exchanger 3)-dependent mechanisms. A PT-selective, mitochondria-targeting adenoviral construct encoding Ad-sglt2-mito-ECFP/Ang II was used to test the hypotheses. The expression of mito-ECFP/Ang II was colocalized primarily with Mito-Tracker Red FM in mouse PT cells or with TMRM in kidney PTs. Mito-ECFP/Ang II markedly increased oxygen consumption rate as an index of mitochondrial oxidative response (69.5%; P <0.01) and extracellular acidification rate as an index of mitochondrial glycolytic response (34%; P <0.01). The mito-ECFP/Ang II–induced oxygen consumption rate and extracellular acidification rate responses were blocked by AT 1 blocker losartan ( P <0.01) and a mitochondria-targeting superoxide scavenger mito-TEMPO ( P <0.01). By contrast, the nonselective NO inhibitor L-NAME alone increased, whereas the mitochondria-targeting expression of AT 2 receptors (mito-AT 2 /GFP) attenuated the effects of mito-ECFP/Ang II ( P <0.01). In the kidney, overexpression of mito-ECFP/Ang II in the mitochondria of the PTs increased systolic blood pressure 12±3 mm Hg ( P <0.01), and the response was attenuated in PT-specific PT- Agtr1a −/− and PT- Nhe3 −/− mice ( P <0.01). Conversely, overexpression of AT 2 receptors selectively in the mitochondria of the PTs induced natriuretic responses in PT- Agtr1a −/− and PT- Nhe3 −/− mice ( P <0.01). Taken together, these results provide new evidence for a physiological role of PT mitochondrial Ang II/AT 1a /superoxide/NHE3 and Ang II/AT 2 /NO/NHE3 signaling pathways in maintaining blood pressure homeostasis.


Life Sciences ◽  
2004 ◽  
Vol 75 (14) ◽  
pp. 1747-1759
Author(s):  
Gunnar Nylund ◽  
Marianne Andersson ◽  
Börje Haraldsson ◽  
Dick S Delbro

2022 ◽  
Vol 20 (1) ◽  
Author(s):  
Mengling Wang ◽  
Feng Zeng ◽  
Fengling Ning ◽  
Yinhang Wang ◽  
Shilin Zhou ◽  
...  

Abstract Background and aims Renal fibrosis is the common outcome in all progressive forms of chronic kidney disease. Unfortunately, the pathogenesis of renal fibrosis remains largely unexplored, among which metabolic reprogramming plays an extremely crucial role in the evolution of renal fibrosis. Ceria nanoparticles (CeNP-PEG) with strong ROS scavenging and anti-inflammatory activities have been applied for mitochondrial oxidative stress and inflammatory diseases. The present study aims to determine whether CeNP-PEG has therapeutic value for renal fibrosis. Methods The unilateral ureteral obstructive fibrosis model was used to assess the therapeutic effects in vivo. Transforming growth factor beta1-induced epithelial-to-mesenchymal transition in HK-2 cells was used as the in vitro cell model. The seahorse bioscience X96 extracellular flux analyzer was used to measure the oxygen consumption rate and extracellular acidification rate. Results In the present study, CeNP-PEG treatment significantly ameliorated renal fibrosis by increased E-cadherin protein expression, and decreased α-SMA, Vimentin and Fibronectin expression both in vitro and in vivo. Additionally, CeNP-PEG significantly reduced the ROS formation and improved the levels of mitochondrial ATP. The seahorse analyzer assay demonstrated that the extracellular acidification rate markedly decreased, whereas the oxygen consumption rate markedly increased, in the presence of CeNP-PEG. Furthermore, the mitochondrial membrane potential markedly enhanced, hexokinase 1 and hexokinase 2 expression significantly decreased after treatment with CeNP-PEG. Conclusions CeNP-PEG can block the dysregulated metabolic status and exert protective function on renal fibrosis. This may provide another therapeutic option for renal fibrosis. Graphical Abstract


Sign in / Sign up

Export Citation Format

Share Document