Arrestins were first described as negative regulators of G protein-coupled receptor (GPCR) signaling via G proteins. New data show that the free and receptor-bound arrestins initiate signaling through MAP kinases, which regulate cell death, survival, and proliferation. In particular, both free and receptor- associated arrestin-3 scaffolds ASK1-MKK-JNK cascade, promoting JNK activation. Here we propose to elucidate the structural basis of arrestin-dependent activation of pro-apoptotic JNK family kinases and their activators MKK4/7 using biochemical and biophysical methods. The molecular mechanisms of the assembly of multi-protein signaling complexes (signalosomes) organized by arrestin-3 will be established, and arrestin-3 residues critical for JNK activation will be identified. Based on ths info, arrestin-3 mutants that bind ASK1, MKK and JNK, but do not promote JNK activation will be constructed. The potential of arrestin mutants with dramatically reduced ability to activate JNKs, several of which we already have, to protect cells against insults and prolong their survival will be tested. We have also constructed arrestin-3 mutants that activate JNKs more efficiently than parental wild type arrestin-3. We established the paradigm where arrestin-3-dependent JNK activation plays key role in cell survival. We will test the ability of hyperactive arrestin-3 mutants to facilitate cell death. We showed that inactive mutants tie up JNKs and upstream kinases in unproductive complexes, thereby acting in dominant- negative manner. We will test the potential of these mutants to protect cells and prolong their survival. Molecular toos that specifically increase or block pro-apoptotic signaling have therapeutic potential in disorders associated with excessive cell proliferation (e.g., cancer) or death (e.g., neurodegenerative diseases).

Public Health Relevance

This proposal focuses on the elucidation of the structural basis of arrestin-dependent activation of pro- apoptotic JNK family kinases and their activators MKK4/7 using biochemical and biophysical methods. The potential of arrestin mutants with dramatically reduced ability to activate JNKs, that were constructed based on this info, to protect cells against insults and prolong their survival will be tested, as well as the ability of the mutats that activate JNKs more efficiently than wild type arrestins to facilitate cell death will be also tested. Molecular tools that specifically increase or block pro-apoptotic signaling have therapeutic potential in disorders associated with excessive cell proliferation (e.g., cancer) or death (e.g., neurodegenerative diseases).

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Research Project (R01)
Project #
5R01GM077561-06
Application #
8458058
Study Section
Biophysics of Neural Systems Study Section (BPNS)
Program Officer
Dunsmore, Sarah
Project Start
2006-04-01
Project End
2016-03-31
Budget Start
2013-04-01
Budget End
2014-03-31
Support Year
6
Fiscal Year
2013
Total Cost
$285,133
Indirect Cost
$88,178
Name
Vanderbilt University Medical Center
Department
Pharmacology
Type
Schools of Medicine
DUNS #
004413456
City
Nashville
State
TN
Country
United States
Zip Code
37212
Tso, Shih-Chia; Chen, Qiuyan; Vishnivetskiy, Sergey A et al. (2018) Using two-site binding models to analyze microscale thermophoresis data. Anal Biochem 540-541:64-75
Gurevich, Vsevolod V; Gurevich, Eugenia V (2018) GPCRs and Signal Transducers: Interaction Stoichiometry. Trends Pharmacol Sci 39:672-684
Cleghorn, Whitney M; Bulus, Nada; Kook, Seunghyi et al. (2018) Non-visual arrestins regulate the focal adhesion formation via small GTPases RhoA and Rac1 independently of GPCRs. Cell Signal 42:259-269
Chen, Qiuyan; Iverson, Tina M; Gurevich, Vsevolod V (2018) Structural Basis of Arrestin-Dependent Signal Transduction. Trends Biochem Sci 43:412-423
Zurkovsky, Lilia; Sedaghat, Katayoun; Ahmed, M Rafiuddin et al. (2017) Arrestin-2 and arrestin-3 differentially modulate locomotor responses and sensitization to amphetamine. Neuropharmacology 121:20-29
Chen, Qiuyan; Perry, Nicole A; Vishnivetskiy, Sergey A et al. (2017) Structural basis of arrestin-3 activation and signaling. Nat Commun 8:1427
Wanka, Lizzy; Babilon, Stefanie; Burkert, Kerstin et al. (2017) C-terminal motif of human neuropeptide Y4 receptor determines internalization and arrestin recruitment. Cell Signal 29:233-239
Prokop, Susanne; Perry, Nicole A; Vishnivetskiy, Sergey A et al. (2017) Differential manipulation of arrestin-3 binding to basal and agonist-activated G protein-coupled receptors. Cell Signal 36:98-107
Vishnivetskiy, Sergey A; Lee, Regina J; Zhou, X Edward et al. (2017) Functional role of the three conserved cysteines in the N domain of visual arrestin-1. J Biol Chem 292:12496-12502
Zhu, Lu; AlmaƧa, Joana; Dadi, Prasanna K et al. (2017) ?-arrestin-2 is an essential regulator of pancreatic ?-cell function under physiological and pathophysiological conditions. Nat Commun 8:14295

Showing the most recent 10 out of 82 publications