1
|
Breen MS, White CH, Shekhtman T, Lin K, Looney D, Woelk CH, Kelsoe JR. Lithium-responsive genes and gene networks in bipolar disorder patient-derived lymphoblastoid cell lines. THE PHARMACOGENOMICS JOURNAL 2016; 16:446-53. [PMID: 27401222 DOI: 10.1038/tpj.2016.50] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 04/21/2016] [Accepted: 05/18/2016] [Indexed: 12/25/2022]
Abstract
Lithium (Li) is the mainstay mood stabilizer for the treatment of bipolar disorder (BD), although its mode of action is not yet fully understood nor is it effective in every patient. We sought to elucidate the mechanism of action of Li and to identify surrogate outcome markers that can be used to better understand its therapeutic effects in BD patients classified as good (responders) and poor responders (nonresponders) to Li treatment. To accomplish these goals, RNA-sequencing gene expression profiles of lymphoblastoid cell lines (LCLs) were compared between BD Li responders and nonresponders with healthy controls before and after treatment. Several Li-responsive gene coexpression networks were discovered indicating widespread effects of Li on diverse cellular signaling systems including apoptosis and defense response pathways, protein processing and response to endoplasmic reticulum stress. Individual gene markers were also identified, differing in response to Li between BD responders and nonresponders, involved in processes of cell cycle and nucleotide excision repair that may explain part of the heterogeneity in clinical response to treatment. Results further indicated a Li gene expression signature similar to that observed with clonidine treatment, an α2-adrenoceptor agonist. These findings provide a detailed mechanism of Li in LCLs and highlight putative surrogate outcome markers that may permit for advanced treatment decisions to be made and for facilitating recovery in BD patients.
Collapse
Affiliation(s)
- M S Breen
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - C H White
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - T Shekhtman
- Veterans Administration, San Diego Healthcare System, San Diego, CA, USA.,Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - K Lin
- Department of Affective Disorder, Guangzhou Brain Hospital, Guangzhou Medical University, Guangzhou, China.,Laboratory of Cognition and Emotion, Guangzhou Brain Hospital, Guangzhou Medical University, Guangzhou, China
| | - D Looney
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA.,Veterans Administration, San Diego Healthcare System, San Diego, CA, USA
| | - C H Woelk
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - J R Kelsoe
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA.,Veterans Administration, San Diego Healthcare System, San Diego, CA, USA.,Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
2
|
Abstract
The high rate of non-responders to initial treatment with antidepressants requires subsequent treatment strategies such as augmentation of antidepressants. Clinical guidelines recommend lithium augmentation as a first-line treatment strategy for non-responding depressed patients. The objectives of this review were to discuss the current place of lithium augmentation in the management of treatment-resistant depression and to review novel findings concerning lithium's mechanisms of action. We conducted a comprehensive and critical review of randomized, placebo-controlled trials, controlled and naturalistic comparator studies, and continuation-phase and discontinuation studies of lithium augmentation in major depression. The outcomes of interest were efficacy, factors allowing outcome prediction and results from preclinical studies investigating molecular mechanisms of lithium action. Substantial efficacy of lithium augmentation in the acute treatment of major depression has been demonstrated in more than 30 open-label studies and 10 placebo-controlled trials. In a meta-analysis addressing the efficacy of lithium in 10 randomized, controlled trials, it had a significant positive effect versus placebo, with an odds ratio of 3.11 corresponding to a number-needed-to-treat (NNT) of 5 and a mean response rate of 41.2% (versus 14.4% in the placebo group). The main limitations of these studies were the relatively small numbers of study participants and the fact that most studies included augmentation of tricyclic antidepressants, which are not in widespread use anymore. Evidence from continuation-phase studies is sparse but suggests that lithium augmentation should be maintained in the lithium-antidepressant combination for at least 1 year to prevent early relapses. Concerning outcome prediction, single studies have reported associations of better outcome rates with more severe depressive symptomatology, significant weight loss, psychomotor retardation, a history of more than three major depressive episodes and a family history of major depression. Additionally, one study suggested a predictive role of the -50T/C single nucleotide polymorphism of the glycogen synthase kinase 3 beta (GSK3B) gene in the probability of response to lithium augmentation. With regard to novel mechanisms of action, GABAergic, neurotrophic and genetic effects might explain the effects of lithium augmentation. In conclusion, augmentation of antidepressants with lithium remains a first-line, evidence-based management option for patients with major depression who have not responded adequately to antidepressants. While the mechanisms of action are currently widely studied, further clinical research on the role of lithium potentiation of the current generation of antidepressants is warranted to reinforce its role as a gold-standard treatment for patients who respond inadequately to antidepressants.
Collapse
|
3
|
Kishore BK, Ecelbarger CM. Lithium: a versatile tool for understanding renal physiology. Am J Physiol Renal Physiol 2013; 304:F1139-49. [PMID: 23408166 DOI: 10.1152/ajprenal.00718.2012] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
By virtue of its unique interactions with kidney cells, lithium became an important research tool in renal physiology and pathophysiology. Investigators have uncovered the intricate relationships of lithium with the vasopressin and aldosterone systems, and the membrane channels or transporters regulated by them. While doing so, their work has also led to 1) questioning the role of adenylyl cyclase activity and prostaglandins in lithium-induced suppression of aquaporin-2 gene transcription; 2) unraveling the role of purinergic signaling in lithium-induced polyuria; and 3) highlighting the importance of the epithelial sodium channel (ENaC) in lithium-induced nephrogenic diabetes insipidus (NDI). Lithium-induced remodeling of the collecting duct has the potential to shed new light on collecting duct remodeling in disease conditions, such as diabetes insipidus. The finding that lithium inhibits glycogen synthase kinase-3β (GSK3β) has opened an avenue for studies on the role of GSK3β in urinary concentration, and GSK isoforms in renal development. Finally, proteomic and metabolomic profiling of the kidney and urine in rats treated with lithium is providing insights into how the kidney adapts its metabolism in conditions such as acquired NDI and the multifactorial nature of lithium-induced NDI. This review provides state-of-the-art knowledge of lithium as a versatile tool for understanding the molecular physiology of the kidney, and a comprehensive view of how this tool is challenging some of our long-standing concepts in renal physiology, often with paradigm shifts, and presenting paradoxical situations in renal pathophysiology. In addition, this review points to future directions in research where lithium can lead the renal community.
Collapse
Affiliation(s)
- Bellamkonda K Kishore
- Nephrology Research (151M) VA SLC Health Care System, 500 Foothill Dr, Salt Lake City, UT 84148, USA.
| | | |
Collapse
|
4
|
Bschor T, Ritter D, Winkelmann P, Erbe S, Uhr M, Ising M, Lewitzka U. Lithium monotherapy increases ACTH and cortisol response in the DEX/CRH test in unipolar depressed subjects. A study with 30 treatment-naive patients. PLoS One 2011; 6:e27613. [PMID: 22132117 PMCID: PMC3223180 DOI: 10.1371/journal.pone.0027613] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 10/20/2011] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Distorted activity of the hypothalamic-pituitary-adrenocortical (HPA) system is one of the most robustly documented biological abnormalities in major depression. Lithium is central to the treatment of affective disorders, but little is known about its effects on the HPA system of depressed subjects. OBJECTIVE To assess the effects of lithium monotherapy on the HPA system of patients with major depression by means of the combined DEX/CRH test. METHOD Thirty drug-naive outpatients with major depression (single episode or unipolar recurrent; SCID I- and II-confirmed) were treated with lithium monotherapy for four weeks. The DEX/CRH test was conducted directly before intake of the first lithium tablet and four weeks thereafter. Weekly ratings with the HDRS(21) were used to determine response (≥50% symptom reduction) and remission (HDRS ≤7). RESULTS Lithium levels within the therapeutic range were achieved rapidly. Tolerability was good; no patient terminated the treatment prematurely. Response and remission rates were 50% and 33% respectively. Compared to the DEX/CRH test before the start of the treatment, a considerable and significant increase in all CRH-stimulated ACTH and cortisol parameters could be detected in the second DEX/CRH test. When analysed with particular regard to responders and non-responders, that significant increase was only present in the responders. CONCLUSIONS We were able to demonstrate that lithium leads to a significant activation of the HPA system. This is possibly connected to stimulation of hypothalamic arginine vasoporessin (AVP), to direct intracellular effects of lithium on pituitary cells and to an induction of gene expression. TRIAL REGISTRATION drks-nue.uniklinik-freiburg.de DRKS00003185.
Collapse
Affiliation(s)
- Tom Bschor
- Department of Psychiatry, Schlosspark-Clinic, Berlin, Germany.
| | | | | | | | | | | | | |
Collapse
|
5
|
Kaidanovich-Beilin O, Lipina TV, Takao K, van Eede M, Hattori S, Laliberté C, Khan M, Okamoto K, Chambers JW, Fletcher PJ, MacAulay K, Doble BW, Henkelman M, Miyakawa T, Roder J, Woodgett JR. Abnormalities in brain structure and behavior in GSK-3alpha mutant mice. Mol Brain 2009; 2:35. [PMID: 19925672 PMCID: PMC2785804 DOI: 10.1186/1756-6606-2-35] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Accepted: 11/19/2009] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Glycogen synthase kinase-3 (GSK-3) is a widely expressed and highly conserved serine/threonine protein kinase encoded by two genes that generate two related proteins: GSK-3alpha and GSK-3beta. Mice lacking a functional GSK-3alpha gene were engineered in our laboratory; they are viable and display insulin sensitivity. In this study, we have characterized brain functions of GSK-3alpha KO mice by using a well-established battery of behavioral tests together with neurochemical and neuroanatomical analysis. RESULTS Similar to the previously described behaviours of GSK-3beta(+/-) mice, GSK-3alpha mutants display decreased exploratory activity, decreased immobility time and reduced aggressive behavior. However, genetic inactivation of the GSK-3alpha gene was associated with: decreased locomotion and impaired motor coordination, increased grooming activity, loss of social motivation and novelty; enhanced sensorimotor gating and impaired associated memory and coordination. GSK-3alpha KO mice exhibited a deficit in fear conditioning, however memory formation as assessed by a passive avoidance test was normal, suggesting that the animals are sensitized for active avoidance of a highly aversive stimulus in the fear-conditioning paradigm. Changes in cerebellar structure and function were observed in mutant mice along with a significant decrease of the number and size of Purkinje cells. CONCLUSION Taken together, these data support a role for the GSK-3alpha gene in CNS functioning and possible involvement in the development of psychiatric disorders.
Collapse
|
6
|
Microarray gene expression profiling of mouse brain mRNA in a model of lithium treatment. Psychiatr Genet 2008; 18:64-72. [DOI: 10.1097/ypg.0b013e3282fb0051] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
7
|
Silva R, Mesquita A, Bessa J, Sousa J, Sotiropoulos I, Leão P, Almeida O, Sousa N. Lithium blocks stress-induced changes in depressive-like behavior and hippocampal cell fate: The role of glycogen-synthase-kinase-3β. Neuroscience 2008; 152:656-69. [DOI: 10.1016/j.neuroscience.2007.12.026] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2007] [Revised: 12/06/2007] [Accepted: 12/12/2007] [Indexed: 02/07/2023]
|
8
|
Li H, Zhou Y, Kang L, Ma L. Single and repeated morphine administrations differently regulate expression of N-ethylmaleimide-sensitive factor gene in the rat brain. Neuroreport 2006; 17:71-4. [PMID: 16361953 DOI: 10.1097/01.wnr.0000192731.62417.be] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The present study investigated the potential effects of single and repeated morphine treatments on gene expression of N-ethylmaleimide-sensitive protein, a key regulatory protein of neurotransmitter release and synaptic plasticity. After a single morphine injection (10 mg/kg), N-ethylmaleimide-sensitive factor gene expression increased significantly in the cortex, hippocampus and periaqueductal gray. In contrast, N-ethylmaleimide-sensitive factor gene expression was not changed after repeated morphine exposures in these brain regions. Sixteen hours after cessation of repeated morphine administration, N-ethylmaleimide-sensitive factor mRNA level increased again to a level approximate to that of the single morphine injection group in the hippocampus, periaqueductal gray and cerebral cortex. These findings suggest that the N-ethylmaleimide-sensitive factor may be involved in the maladaptive changes in the nervous system after opiate treatment.
Collapse
Affiliation(s)
- Haohong Li
- Pharmacology Research Center, Department of Pharmacology, Shanghai Medical College Fudan University, Shanghai, China
| | | | | | | |
Collapse
|
9
|
de Vasconcellos APS, Zugno AI, Dos Santos AHDP, Nietto FB, Crema LM, Gonçalves M, Franzon R, de Souza Wyse AT, da Rocha ER, Dalmaz C. Na+,K(+)-ATPase activity is reduced in hippocampus of rats submitted to an experimental model of depression: effect of chronic lithium treatment and possible involvement in learning deficits. Neurobiol Learn Mem 2005; 84:102-10. [PMID: 15961330 DOI: 10.1016/j.nlm.2005.05.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2005] [Revised: 05/03/2005] [Accepted: 05/05/2005] [Indexed: 11/26/2022]
Abstract
This study was undertaken to verify the effects of chronic stress and lithium treatments on the hippocampal Na+,K(+)-ATPase activity of rats, as well as to investigate the effects of stress interruption and post-stress lithium treatment on this enzyme activity and on spatial memory. Two experiments were carried out; in the first experiment, adult male Wistar rats were divided into two groups: control and submitted to a chronic variate stress paradigm, and subdivided into treated or not with LiCl. After 40 days of treatment, rats were killed, and Na+,K(+)-ATPase activity was determined. In the second experiment, rats were stressed during 40 days, and their performance was evaluated in the Water Maze task. The stressed group was then subdivided into four groups, with continued or interrupted stress treatment and treated or not with lithium for 30 additional days. After a second evaluation of performance in the Water Maze, rats were killed and Na+,K(+)-ATPase activity was also measured. Results showed an impairment in Na+,K(+)-ATPase activity and in Water Maze performance of chronically stressed rats, which were prevented by lithium treatment and reversed by lithium treatment and by stress interruption. These results suggest that the modulation of Na+,K(+)-ATPase activity may be one of the mechanisms of action of lithium in the treatment of mood disorders.
Collapse
|
10
|
McQuade R, Leitch MM, Gartside SE, Young AH. Effect of chronic lithium treatment on glucocorticoid and 5-HT1A receptor messenger RNA in hippocampal and dorsal raphe nucleus regions of the rat brain. J Psychopharmacol 2004; 18:496-501. [PMID: 15582915 DOI: 10.1177/026988110401800406] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The therapeutic mechanism of action of lithium in the treatment of bipolar disorder is not well understood. Dysfunction of both 5-HT(1A) receptor mediated neurotransmission and the glucocorticoid receptor is associated with mood disorders, and preclinical studies suggest that lithium treatment can modulate these receptor subtypes. In this study, we investigated the effect of chronic lithium treatment on 5-HT(1A) receptors and glucocorticoid receptors in the rat brain. Male Sprague-Dawley rats were treated with lithium (3 mmol/kg/day) or saline for 28 days via subcutaneous implanted mini-osmotic pumps. After 28 days of treatment, the expression of mRNA for 5-HT(1A) receptors and glucocorticoid receptors in the rat hippocampus and dorsal raphe nucleus was determined by in situ hybridization histochemistry. Chronic administration of lithium decreased mRNA coding for post-synaptic 5-HT(1A) receptors in hippocampal subregions but not for somatodentritic 5-HT(1A) receptors in the dorsal raphe nucleus. Chronic administration of lithium did not affect mRNA coding for glucocorticoid receptors in hippocampal subregions or in the dorsal raphe nucleus. Mean plasma lithium levels in the lithium-treated group were 0.50 +/- 0.03 mmol/l; all animals appeared healthy and maintained a normal increase in body weight. Given recent reports implicating hypersensitive post-synaptic 5-HT(1A) receptors in bipolar manic patients, the present study suggests that down-regulation of this receptor population may be important in the therapeutic mechanism of action of lithium.
Collapse
Affiliation(s)
- Richard McQuade
- Psychobiology Research Group, School of Neurology, Neurobiology and Psychiatry, The Medical School, Newcastle Upon Tyne NE2 4HH, UK.
| | | | | | | |
Collapse
|
11
|
Semba J, Akanuma N, Wakuta M, Tanaka N, Suhara T. Alterations in the expressions of mRNA for GDNF and its receptors in the ventral midbrain of rats exposed to subchronic phencyclidine. ACTA ACUST UNITED AC 2004; 124:88-95. [PMID: 15093689 DOI: 10.1016/j.molbrainres.2004.02.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2004] [Indexed: 11/18/2022]
Abstract
Phencyclidine (PCP) produces schizophrenia-like symptoms in normal humans. This suggests that the dysfunction of glutamatergic neurotransmission may play an important role in the pathology of schizophrenia. However, PCP also exerts its effect on the mesolimbic dopamine (DA) system and modulates DA function in the brain, the abnormality of which is proposed to be a main pathology of schizophrenia. Recently, glial cell-line derived neurotrophic factor (GDNF) has been shown to play a protective role for DA neurons against neurotoxic injuries and maintaining DA function in the brain. We hypothesized that subchronic PCP may alter the function of GDNF in the ventral midbrain, where DA cell bodies are localized. Male Wistar rats were injected intraperitoneally with PCP daily for 10 days at 5 or 10 mg/kg, and their brains were removed 24 h after the last injection. The expressions of GDNF and its receptor (GFRalpha-1 and c-ret) mRNAs in the substantia nigra compacta (SNC) and ventral tegmental area (VTA) were determined by non-radioactive in situ hybridization, and those of GDNF and c-ret mRNA were found to be increased after the PCP subchronic administration. No significant changes, however, were observed in the expressions of GFRalpha-1 and basic fibroblast growth factor. These results suggest that subchronic PCP may modulate the function of the GDNF system, which exerts a trophic action on DA neurons in the ventral midbrain.
Collapse
Affiliation(s)
- Jun'ichi Semba
- Division of Health Sciences, University of the Air, Wakaba, Mihama, Chiba, Japan.
| | | | | | | | | |
Collapse
|
12
|
Vasconcellos APS, Tabajara AS, Ferrari C, Rocha E, Dalmaz C. Effect of chronic stress on spatial memory in rats is attenuated by lithium treatment. Physiol Behav 2003; 79:143-9. [PMID: 12834784 DOI: 10.1016/s0031-9384(03)00113-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Stress is known to alter cognitive functions, such as memory, and it has been linked to the pathophysiology of mood and anxiety disorders. Chronic lithium treatment is used in some psychiatric disorders and has been suggested to act upon mechanisms which can enhance neuronal viability. The purpose of this work is to investigate a possible effect of lithium treatment in a chronic stress model. Adult male Wistar rats were divided in two groups, control and chronically stressed, treated either with normal chow or with chow containing LiCl for 40 days. Stress treatment was a chronic variable stress model, consisting of different stressors which were applied in a random fashion, once a day, every day. Memory was assessed by using the water maze task. The results demonstrated a marked decrease in reference memory in the water maze task in chronically stressed rats. This effect was attenuated by lithium treatment in all the parameters considered. No effect was observed in the working memory. These results indicate that lithium treatment may counteract some effects of chronic stress situations, particularly concerning spatial memory.
Collapse
Affiliation(s)
- A P S Vasconcellos
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Ramiro Barcelos, 2600 (Anexo) Lab. 32, 90035-003, RS, Porto Alegre, Brazil.
| | | | | | | | | |
Collapse
|
13
|
Fan XL, Zhang JS, Zhang XQ, Yue W, Ma L. Differential regulation of beta-arrestin 1 and beta-arrestin 2 gene expression in rat brain by morphine. Neuroscience 2003; 117:383-9. [PMID: 12614678 DOI: 10.1016/s0306-4522(02)00930-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Beta-arrestins are a family of regulatory and scaffold proteins functioning in signal transduction of G protein-coupled receptors including opioid receptors. Upon agonist stimulation, beta-arrestins bind to opioid receptors phosphorylated by G protein-coupled receptor kinases and promote receptor internalization and desensitization. Studies indicated that beta-arrestins are required in the development of morphine tolerance in mice. In the current study, we investigated the potential regulatory effects of morphine administration on beta-arrestin 1 and beta-arrestin 2 mRNA levels in different brain regions in rat using in situ hybridization method. Our results showed that the acute morphine administration (10 mg/kg) resulted in approximately 30% reduction in both beta-arrestin 1 and beta-arrestin 2 mRNA levels in hippocampus while the chronic morphine treatment (10 mg/kg, b.i.d., for 9 days) caused no significant change in level of either beta-arrestin mRNA. In locus coeruleus, both acute and chronic morphine treatments resulted in significant decreases (over 50%) in beta-arrestin 1 mRNA level but failed to induce any change in the level of beta-arrestin 2 gene expression. The acute morphine administration had no significant effect on beta-arrestin 1 or beta-arrestin 2 mRNA level in periaqueductal gray and cerebral cortex. However, after chronic morphine treatment, beta-arrestin 2 mRNA level decreased by 40% in periaqueductal gray and increased by 25% in cerebral cortex, in strong contrast to the unchanged beta-arrestin 1 mRNA level in these two brain regions. Furthermore, spontaneous or naloxone-precipitated withdrawal of morphine that did not affect the level of beta-arrestin 1 mRNA resulted in an aberrant increase (100% over control) in beta-arrestin 2 mRNA level in hippocampus. Our results thus demonstrated for the first time that opiate administration regulates level of beta-arrestin mRNAs in brain and the expression of beta-arrestin 1 and beta-arrestin 2 subtypes is differentially regulated in locus coeruleus, periaqueductal gray, and cerebral cortex by morphine. These data suggest that beta-arrestin 1 and beta-arrestin 2 may play different roles in the development of opioid tolerance and dependence.
Collapse
Affiliation(s)
- X L Fan
- National Laboratory of Medical Neurobiology, Fudan University Medical Center, 138 Yi Xue Yuan Road, Shanghai 200032, People's Republic of China
| | | | | | | | | |
Collapse
|
14
|
Fan XL, Zhang JS, Zhang XQ, Ma L. Chronic morphine treatment and withdrawal induce up-regulation of c-jun n-terminal kinase 3 gene expression in rat brain. Neuroscience 2003; 122:997-1002. [PMID: 14643766 DOI: 10.1016/j.neuroscience.2003.08.062] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Chronic opiate applications produce long-term impacts on many functions of the brain and induce tolerance, dependence, and addiction. It has been demonstrated that opioid drugs are capable to induce apoptosis of neuronal cells, but the mechanism is not clear. c-Jun N-terminal kinase 3 (JNK3), specifically expressed in brain, has been proved to mediate neuronal apoptosis and is involved in opiate-induced cell apoptosis in vitro. The present study investigated the effect of opioid administration on expression of JNK3, an important mediator involved in apoptosis of neurons, in rat brain. Our results showed that single or chronic injection of morphine resulted in a 45-50% increase in the level of JNK3 mRNA in frontal cortex, while no significant change was detected in other brain regions such as thalamus, hippocampus and locus coeruleus. Similar to what was observed after the acute or chronic morphine administration, no significant change in JNK3 expression was detected in locus coeruleus following cessation of the chronic morphine administration. However, interestingly, sustained elevation of JNK3 expression peaked on day 14 after cessation of morphine treatment was observed in the brain regions such as hippocampus and thalamus, where acute or chronic morphine treatment did not cause any significant change in JNK3 gene expression. The increased JNK3 mRNA in these brain areas returned to the control levels in 28 days following cessation of chronic morphine treatment. Taken together, these results demonstrated for the first time that the expression of JNK3 gene is regulated by opioids and that chronic opioid administration and withdrawal could induce sustained elevation of JNK3 mRNA in many important brain areas. The changes in JNK3 gene expression in brain induced by chronic opioid treatment may play a role in opioid-induced apoptosis and neurotoxicity.
Collapse
Affiliation(s)
- X-L Fan
- National Laboratory of Medical Neurobiology, Shanghai Medical College, Fudan University, 138 Yi Xue Yuan Road, Shanghai 200032, People's Republic of China
| | | | | | | |
Collapse
|
15
|
Fan X, Zhang J, Zhang X, Yue W, Ma L. Acute and chronic morphine treatments and morphine withdrawal differentially regulate GRK2 and GRK5 gene expression in rat brain. Neuropharmacology 2002; 43:809-16. [PMID: 12384166 DOI: 10.1016/s0028-3908(02)00147-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Opioid agonist stimulates activation of G protein-coupled receptor kinase (GRK) and causes desensitization of opioid signaling, which plays an important role in opioid tolerance. The current study investigated the potential regulatory effects of acute and chronic morphine administration and withdrawal on GRK2 and GRK5 gene expression in rat brain. Our results showed that the initial morphine treatment (10 mg/kg) significantly increased GRK mRNA levels in cerebral cortex, hippocampus, and lateral thalamic nuclei. A significant decrease in GRK5 mRNA levels was observed in periaqueductal gray. In strong contrast, repeated administration of morphine for 9 days failed to cause any significant increase in GRK5 mRNA in any of these brain regions. Chronic morphine treatment resulted in 30-70% down-regulation of GRK2 expression in cerebral cortex, hippocampus, thalamus, and locus coeruleus, opposite to what observed with the single morphine administration. Moreover, spontaneous and naloxone-precipitated morphine withdrawal resulted in aberrant increases in GRK2 and GRK5 mRNA levels in these brain regions. Taken together, our study suggests that opioid not only induces rapid negative feedback regulation on opioid signals through activation of GRK but also exerts its impact, via controlling levels of GRK gene expression, on the regulatory machinery itself over a longer period of time in brain.
Collapse
Affiliation(s)
- X Fan
- National Laboratory of Medical Neurobiology, Fudan University Medical Center, 138 Yi Xue Yuan Road, Shanghai 200032, People's Republic of China
| | | | | | | | | |
Collapse
|