2013
DOI: 10.1074/jbc.m112.436006
|View full text |Cite
|
Sign up to set email alerts
|

Aminothienopyridazines and Methylene Blue Affect Tau Fibrillization via Cysteine Oxidation

Abstract: Background: Tau fibrillization inhibitors hold promise as potential therapeutic agents for neurodegenerative disease. Results: The Tau fibrillization inhibitors, aminothienopyridazines and methylene blue, promote disulfide bond formation in Tau. Conclusion: These compounds affect Tau fibrillization by a relatively nonspecific oxidative mechanism. Significance: Understanding the mechanism by which these compounds affect Tau fibril formation provides insights into their potential as therapeutic agents.

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
1
1

Citation Types

2
139
3

Year Published

2013
2013
2022
2022

Publication Types

Select...
7
2

Relationship

0
9

Authors

Journals

citations
Cited by 133 publications
(144 citation statements)
references
References 52 publications
(86 reference statements)
2
139
3
Order By: Relevance
“…Some compounds foster this reaction when incubated with Tau under non-reducing conditions (21). To assess the contribution of Cys residues to cyanine activity, the interaction of 2N4R C291A,C322A…”
Section: Cyanine-mediated Oligomer Formation Requires Inducer and Thementioning
confidence: 99%
See 1 more Smart Citation
“…Some compounds foster this reaction when incubated with Tau under non-reducing conditions (21). To assess the contribution of Cys residues to cyanine activity, the interaction of 2N4R C291A,C322A…”
Section: Cyanine-mediated Oligomer Formation Requires Inducer and Thementioning
confidence: 99%
“…For example, aldehydes can covalently modify Tau protein monomer, thereby trapping it in aggregation incompetent forms (19). Other compounds promote Cys oxidation under non-reducing conditions and extended incubation times, again yielding assembly incompetent monomer (20,21). Neither of these covalent mechanisms is predicted to have utility in vivo.…”
mentioning
confidence: 99%
“…These compoundscould interfere in readouts by multiple ways including: nonselective reactivity with proteins [2] fluorescence [3], cysteine oxidation [4], aggregation [5], redox activity [6], membrane disruption [7] and chelation [8] among others.PAINS are commonly reported in the literature as promising hits against different proteins; however, an accurate look in chemical structure can help biochemists and pharmacologists to identify these structures before testing them.…”
mentioning
confidence: 99%
“…[4] Other redox-active compounds, including the non-neuroleptic phenothiazine methylene blue (MB) [methylthioninium chloride (MTC), Rember™, TRx-0014, TauRx Therapeutics, Singapore, Republic of Singapore] can also modulate cysteine oxidation when incubated in the absence of exogenous reducing agents. [7] In general, covalent mechanisms of tau-aggregation inhibition in AD are predicted to have low utility in vivo. [8] However, dimethylfumarate, an electrophile capable of reacting covalently with cysteine sulfhydryls, was approved for oral treatment of multiple sclerosis, [9] suggesting that electrophilic compounds acting through covalent inhibitory mechanisms can be useful therapeutic agents.…”
Section: Introductionmentioning
confidence: 99%