2005
DOI: 10.1016/j.molbrainres.2005.04.004
|View full text |Cite
|
Sign up to set email alerts
|

Chronic morphine acts via a protein kinase Cγ–Gβ–adenylyl cyclase complex to augment phosphorylation of Gβ and Gβγ stimulatory adenylyl cyclase signaling

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
1
1

Citation Types

4
36
0

Year Published

2007
2007
2020
2020

Publication Types

Select...
7
1

Relationship

1
7

Authors

Journals

citations
Cited by 31 publications
(40 citation statements)
references
References 42 publications
4
36
0
Order By: Relevance
“…We hypothesized that the strategies that cells use to rescue themselves from the sustained activation of opioid receptors would depend on the consequences of their short-term activation in the opioid-naive state. This formulation predicts that previously demonstrated sequelae of longterm morphine treatment [e.g., augmented phosphorylation of AC (Chakrabarti et al, 1998b) and the G␤ subunit of G proteins (Chakrabarti et al, 2001(Chakrabarti et al, , 2005b …”
Section: Introductionmentioning
confidence: 53%
See 2 more Smart Citations
“…We hypothesized that the strategies that cells use to rescue themselves from the sustained activation of opioid receptors would depend on the consequences of their short-term activation in the opioid-naive state. This formulation predicts that previously demonstrated sequelae of longterm morphine treatment [e.g., augmented phosphorylation of AC (Chakrabarti et al, 1998b) and the G␤ subunit of G proteins (Chakrabarti et al, 2001(Chakrabarti et al, , 2005b …”
Section: Introductionmentioning
confidence: 53%
“…We hypothesized that the strategies that cells use to rescue themselves from the sustained activation of opioid receptors would depend on the consequences of their short-term activation in the opioid-naive state. This formulation predicts that previously demonstrated sequelae of longterm morphine treatment [e.g., augmented phosphorylation of AC (Chakrabarti et al, 1998b) and the G␤ subunit of G proteins (Chakrabarti et al, 2001(Chakrabarti et al, , 2005bChakrabarti and Gintzler, 2003], increased membrane translocation of protein kinase C␥ (PKC␥) Zeitz et al, 2001;Chakrabarti et al, 2005b), increased MOR G␣ s association (Chakrabarti et al, 2005a), all of which shift short-term MOR-coupled signaling from AC inhibitory to stimulatory (Gintzler and Chakrabarti, 2006), are not hard-wired and in fact will be negated, or reversed, in opioid-naive cells manifesting short-term stimulatory responsiveness to MOR activation.…”
mentioning
confidence: 53%
See 1 more Smart Citation
“…GluA4 can also be phosphorylated at Ser842 by PKC-g (Gomes et al, 2007;Zheng and Keifer, 2008). Deletion of the gene encoding PKC-g abolishes opioid-induced hypersensitivity (Célérier et al, 2004), whereas repeated administration of m-opioid agonists increases PKC-g activity (Chakrabarti et al, 2005;Narita et al, 2001). Thus, PKC-g may represent an additional key player in the regulation of trafficking of GluA4-containing AMPAR by opioids.…”
Section: Discussionmentioning
confidence: 99%
“…6,23,28,30,32,51,54,65,71 Prolonged opioid treatment has also been shown to alter the function of Gβγ-subunits of G proteins coupling to MORs, resulting in multiplicative excitatory effects such as phosphorylation of Gβ and enhanced Gβγ stimulation of adenylyl cyclase, phosphorylation of G protein receptor kinase 2/3 and Gβ via protein kinase C, and increased activity of PKA, which can also be activated by G s stimulation of adenylyl cyclase. [15][16][17][18][19][20]37,57 The Gβγ that associates with adenylyl cyclases during opioid tolerance has now been shown to originate from the G s protein coupling to MOR. 72 In conjunction with increased excitatory signaling via MOR-associated Gα s in an injury state, the Gβγ released by these G s heterotrimeric proteins may further enhance the analgesic tolerance and hyperalgesia observed clinically.…”
Section: Discussionmentioning
confidence: 99%