Extrapyramidal Motor Side Effects

Author(s):  
Bart Ellenbroek ◽  
Alfonso Abizaid ◽  
Shimon Amir ◽  
Martina de Zwaan ◽  
Sarah Parylak ◽  
...  
1979 ◽  
Vol 7 (1) ◽  
pp. 19-28 ◽  
Author(s):  
H Hökendorf

An electromyographic method was used to study the effect of combination therapy with L-dopa and trihexyphenidyl on tremor in thirty patients suffering from extrapyramidal motor system disease. In this method tremor activity was measured and documented so that the course of the disease could be followed objectively. L-dopa alone was slightly more effective against tremor than was trihexyphenidyl alone. The combination of the two drugs was more effective than either drug used alone, and its side-effects were mild and definitely fewer than had been reported with L-dopa combined with a decarboxylase inhibitor. Good control of tremor with L-dopa and trihexyphenidyl was obtained clinically and verified electromyographically.


2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Uri Nimrod Ramírez-Jarquín ◽  
Neelam Shahani ◽  
William Pryor ◽  
Alessandro Usiello ◽  
Srinivasa Subramaniam

Abstract The mammalian target of rapamycin (mTOR) is a ubiquitously expressed serine/threonine kinase protein complex (mTORC1 or mTORC2) that orchestrates diverse functions ranging from embryonic development to aging. However, its brain tissue-specific roles remain less explored. Here, we have identified that the depletion of the mTOR gene in the mice striatum completely prevented the extrapyramidal motor side effects (catalepsy) induced by the dopamine 2 receptor (D2R) antagonist haloperidol, which is the most widely used typical antipsychotic drug. Conversely, a lack of striatal mTOR in mice did not affect catalepsy triggered by the dopamine 1 receptor (D1R) antagonist SCH23390. Along with the lack of cataleptic effects, the administration of haloperidol in mTOR mutants failed to increase striatal phosphorylation levels of ribosomal protein pS6 (S235/236) as seen in control animals. To confirm the observations of the genetic approach, we used a pharmacological method and determined that the mTORC1 inhibitor rapamycin has a profound influence upon post-synaptic D2R-dependent functions. We consistently found that pretreatment with rapamycin entirely prevented (in a time-dependent manner) the haloperidol-induced catalepsy, and pS6K (T389) and pS6 (S235/236) signaling upregulation, in wild-type mice. Collectively, our data indicate that striatal mTORC1 blockade may offer therapeutic benefits with regard to the prevention of D2R-dependent extrapyramidal motor side effects of haloperidol in psychiatric illness.


2020 ◽  
Author(s):  
Uri Nimrod Ramírez-Jarquín ◽  
Neelam Shahani ◽  
William Pryor ◽  
Alessandro Usiello ◽  
Srinivasa Subramaniam

ABSTRACTThe mammalian target of rapamycin (mTOR) is a ubiquitously expressed serine/threonine kinase protein complex (mTORC1 or mTORC2) that orchestrates diverse functions ranging from embryonic development to aging. However, its brain tissue-specific roles remain less explored. Here, we have identified that the depletion of the mTOR gene in the mice striatum completely prevented the extrapyramidal motor side-effects (catalepsy) induced by the dopamine 2 receptor (D2R) antagonist haloperidol, which is the most widely used typical antipsychotic drug. Conversely, a lack of striatal mTOR in mice did not affect catalepsy triggered by the dopamine 1 receptor (D1R) antagonist SCH23390. Along with the lack of cataleptic effects, the administration of haloperidol in mTOR mutants failed to increase striatal phosphorylation levels of ribosomal protein pS6 (S235/236) as seen in control animals. To confirm the observations of the genetic approach, we used a pharmacological method and determined that the mTORC1 inhibitor rapamycin has a profound influence upon post-synaptic D2R-dependent functions. We consistently found that pretreatment with rapamycin entirely prevented (in a time-dependent manner) the haloperidol-induced catalepsy in wild-type mice. Collectively, our data indicate that striatal mTORC1 blockade may offer therapeutic benefits with regard to the prevention of D2R-dependent extrapyramidal motor side-effects of haloperidol in psychiatric illness.


1986 ◽  
Vol 19 (04) ◽  
pp. 149-151 ◽  
Author(s):  
A. Christensen ◽  
W. Greil ◽  
H. Haag ◽  
U. Havemann ◽  
W. Kehr ◽  
...  

2010 ◽  
Vol 25 ◽  
pp. 64
Author(s):  
R. Musil ◽  
D. Rujescu ◽  
I. Spellmann ◽  
A. Mayr ◽  
P. Zill ◽  
...  

2022 ◽  
Vol 12 (1) ◽  
Author(s):  
Chuanjun Zhuo ◽  
Yong Xu ◽  
Weihong Hou ◽  
Jiayue Chen ◽  
Qianchen Li ◽  
...  

AbstractAntipsychotic pharmacotherapy has been widely recommended as the standard of care for the treatment of acute schizophrenia and psychotic symptoms of other psychiatric disorders. However, there are growing concerns regarding antipsychotic-induced side effects, including weight gain, metabolic syndrome (MetS), and extrapyramidal motor disorders, which not only decrease patient compliance, but also predispose to diabetes and cardiovascular diseases. To date, most studies and reviews on the mechanisms of antipsychotic-induced metabolic side effects have focused on central nervous system mediation of appetite and food intake. However, disturbance in glucose and lipid metabolism, and hepatic steatosis induced by antipsychotic drugs might precede weight gain and MetS. Recent studies have demonstrated that the mechanistic/mammalian target of rapamycin (mTOR) pathway plays a critical regulatory role in the pathophysiology of antipsychotic drug-induced disorders of hepatic glucose and lipid metabolism. Furthermore, antipsychotic drugs promote striatal mTOR pathway activation that contributes to extrapyramidal motor side effects. Although recent findings have advanced the understanding of the role of the mTOR pathway in antipsychotic-induced side effects, few reviews have been conducted on this emerging topic. In this review, we synthesize key findings by focusing on the roles of the hepatic and striatal mTOR pathways in the pathogenesis of metabolic and extrapyramidal side effects, respectively. We further discuss the potential therapeutic benefits of normalizing excessive mTOR pathway activation with mTOR specific inhibitors. A deeper understanding of pathogenesis may inform future intervention strategies using the pharmacological or genetic inhibitors of mTOR to prevent and manage antipsychotic-induced side effects.


Sign in / Sign up

Export Citation Format

Share Document