2004
DOI: 10.1016/j.bmcl.2003.11.077
|View full text |Cite
|
Sign up to set email alerts
|

N -[1-Aryl-2-(1-imidazolo)ethyl]-guanidine derivatives as potent inhibitors of the bovine mitochondrial F 1 F 0 ATP hydrolase

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
3
1
1

Citation Types

0
15
0

Year Published

2004
2004
2021
2021

Publication Types

Select...
7
2

Relationship

0
9

Authors

Journals

citations
Cited by 15 publications
(15 citation statements)
references
References 21 publications
0
15
0
Order By: Relevance
“…Except for the mitochondrial inhibitor protein of ATP hydrolysis, IF 1 [55], we were only able to find one extant report of any other such inhibitor [56], with the compounds described being a series of substituted guanidine derivatives.…”
Section: Discussionmentioning
confidence: 99%
“…Except for the mitochondrial inhibitor protein of ATP hydrolysis, IF 1 [55], we were only able to find one extant report of any other such inhibitor [56], with the compounds described being a series of substituted guanidine derivatives.…”
Section: Discussionmentioning
confidence: 99%
“…For example, during cardiac ischemia, cardioprotective benefit is thought to derive from preventing the destruction of ATP that leads to tissue damage by inhibiting the hydrolytic activity of pyran, guanidine, and benzodiazapines (69)(70)(71). Similarly, oligomycin, which inhibits F 1 F o -ATPase through its F o domain, preserves ATP and protects against or postpones injury during ischemia (72).…”
Section: Mechanism Of Inhibitionmentioning
confidence: 99%
“…The toxicological modulation of F0-ATP hydrolase-based inhibition may provide a novel mechanism to prevent different pathological conditions where mitochondrial ATP-hydrolysis mechanisms are exacerbated. Recent experimental evidence suggest that the mitochondrial Oligomycin A-sensitive F0-ATPase subunit is an uncoupling channel within the mitochondrial permeability transition pore, which is responsible for inducing mitochondrial dysfunction linked to apoptosis. This in turn can thus be an interesting mitotoxic-targeting therapy based on the F0-ATP hydrolase inhibition under pathological conditions, as has been suggested in previous investigations using experimental in vitro assays combined with traditional approach-based quantitative structure–activity relationship models (SAR/QSAR). ,,, …”
Section: Introductionmentioning
confidence: 97%
“…The H + proton flux through the membrane-embedded F0-ATPsynthase subunit of mitochondrial F0F1-ATP synthase favors mechanical rotation coupled to the γ and the ε subunits co-rotating within the F1-ATP synthase subunit, which induces synchronized conformational changes resulting in the synthesis and release of ATP. However, under pathological conditions like cancer, heart ischemia/reperfusion, cerebrovascular events, and mitochondrial encephalomyopathy, the membrane-embedded F0-ATPsynthase subunit paradoxically hydrolyzes ATP associated with the reverse biochemical reaction consuming the ATP reserves and quickly compromising the cellular homeostasis and viability …”
Section: Introductionmentioning
confidence: 99%