1
|
Prescribing Tamoxifen in Patients With Mood Disorders: A Systematic Review of Potential Antimanic Versus Depressive Effects. J Clin Psychopharmacol 2021; 41:450-460. [PMID: 34166298 DOI: 10.1097/jcp.0000000000001412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE/BACKGROUND Tamoxifen is a selective estrogen receptor modulator widely used for treatment and prevention of estrogenic receptor-positive breast cancer. Tamoxifen is an object of growing interest in psychopharmacology as an antimanic drug, because it inhibits the protein kinase C, a molecular target of bipolar disorder. Consistently, the potential depressive effect of tamoxifen has been repeatedly reported. METHODS/PROCEDURES This article systematically reviews studies examining tamoxifen impact on mood, exploring either its potential therapeutic use as antimanic agent or its potential depressive effect. FINDINGS Eight studies explored tamoxifen antimanic properties, all, but one, reported a rapid and efficacious antimanic action. As to the depressive effect, 9 cohort studies emerged among which 4 pointed out an increased risk of depression. Seven case reports described the onset or exacerbation of depressive episodes besides 1 case series study reported a high rate of depressive symptoms. In addition, 1 case report study described a tamoxifen-induced manic episode. IMPLICATIONS/CONCLUSIONS The present review highlights tamoxifen treatment as a possible trigger of mood symptoms onset or exacerbation in vulnerable patients. Accordingly, patients with a history of mood disorders may require a close clinical surveillance during tamoxifen use. At the same time, the use of tamoxifen as an antimanic agent in psychiatric settings requires caution, as available evidence came from small-sample studies with short observation time. More studies are needed to define how long-term tamoxifen use may affect the course of bipolar disorder.
Collapse
|
2
|
Zheng Y, Yang C, Zheng X, Guan Q, Yu S. Acrylamide treatment alters the level of Ca 2+ and Ca 2+-related protein kinase in spinal cords of rats. Toxicol Ind Health 2021; 37:113-123. [PMID: 33487136 DOI: 10.1177/0748233720971879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This study aimed to analyze the neurological changes induced by acrylamide (ACR) poisoning and their underlying mechanisms within the spinal cords of male adult Wistar rats. The rats were randomly divided into three groups (n = 9 rats per group). ACR was intraperitoneally injected to produce axonopathy according to the daily dosing schedules of 20 or 40 mg/kg/day of ACR for eight continuous weeks (three times per week). During the exposure period, body weights and gait scores were assessed, and the concentration of Ca2+ was calculated in 27 mice. Protein kinase A (PKA), protein kinase C (PKC), cyclin-dependent protein kinase 5 (CDK5), and P35 were assessed by electrophoretic resolution and Western blotting. The contents of 3'-cyclic adenosine monophosphate (cAMP) and calmodulin (CaM) were determined using ELISA kits, and the activities of calcium/calmodulin-dependent protein kinase II (CaMKII), PKA, and PKC were determined using the commercial Signa TECTPKAassay kits. Compared with control rats, treatment with 20 and 40 mg/kg of ACR decreased body weight and increased gait scores at 8 weeks. Intracellular Ca2+ levels increased significantly in treated rats; CaM, PKC, CDK5, and P35 levels were significantly decreased; and PKA and cAMP levels remained unchanged. CaMKII, PKA, and PKC activities increased significantly. The results indicated that ACR can damage neurofilaments by affecting the contents and activities of CaM, CaMKII, PKA, cAMP, PKC, CDK5, and P35, which could result in ACR toxic neuropathy.
Collapse
Affiliation(s)
- Yunhe Zheng
- Gansu Provincial Center for Disease Control and Prevention, West Lanzhou, Gansu, People's Republic of China
| | - Chen Yang
- Gansu Provincial Center for Disease Control and Prevention, West Lanzhou, Gansu, People's Republic of China
| | - Xiu'e Zheng
- Shandong food and Drug Administration, SDFDA, Jinan, People's Republic of China
| | - Qiangdong Guan
- School of Public Health, Nanjing Medical University, Jiangning District, Nanjing, People's Republic of China
| | - Sufang Yu
- School of Public Health, 12589Shandong University, Jinan, People's Republic of China
| |
Collapse
|
3
|
Li CY, Ma W, Liu KP, Yang JW, Wang XB, Wu Z, Zhang T, Wang JW, Liu W, Liu J, Liang Y, Zhang XK, Li JJ, Guo JH, Li LY. Advances in intervention methods and brain protection mechanisms of in situ and remote ischemic postconditioning. Metab Brain Dis 2021; 36:53-65. [PMID: 33044640 DOI: 10.1007/s11011-020-00562-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/05/2020] [Indexed: 01/01/2023]
Abstract
Ischemic postconditioning (PostC) conventionally refers to a series of brief blood vessel occlusions and reperfusions, which can induce an endogenous neuroprotective effect and reduce cerebral ischemia/reperfusion (I/R) injury. Depending on the site of adaptive ischemic intervention, PostC can be classified as in situ ischemic postconditioning (ISPostC) and remote ischemic postconditioning (RIPostC). Many studies have shown that ISPostC and RIPostC can reduce cerebral IS injury through protective mechanisms that increase cerebral blood flow after reperfusion, decrease antioxidant stress and anti-neuronal apoptosis, reduce brain edema, and regulate autophagy as well as Akt, MAPK, PKC, and KATP channel cell signaling pathways. However, few studies have compared the intervention methods, protective mechanisms, and cell signaling pathways of ISPostC and RIPostC interventions. Thus, in this article, we compare the history, common intervention methods, neuroprotective mechanisms, and cell signaling pathways of ISPostC and RIPostC.
Collapse
Affiliation(s)
- Chun-Yan Li
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Wei Ma
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Kuang-Pin Liu
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Jin-Wei Yang
- Second Department of General Surgery, First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China
| | - Xian-Bin Wang
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Zhen Wu
- Second Department of General Surgery, First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China
| | - Tong Zhang
- Second Department of General Surgery, First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China
| | - Jia-Wei Wang
- Second Department of General Surgery, First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China
| | - Wei Liu
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Jie Liu
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Yu Liang
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Xing-Kui Zhang
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Jun-Jun Li
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Jian-Hui Guo
- Second Department of General Surgery, First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China.
| | - Li-Yan Li
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China.
| |
Collapse
|
4
|
Horbach T, Götz C, Kietzmann T, Dimova EY. Protein kinases as switches for the function of upstream stimulatory factors: implications for tissue injury and cancer. Front Pharmacol 2015; 6:3. [PMID: 25741280 PMCID: PMC4332324 DOI: 10.3389/fphar.2015.00003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 01/07/2015] [Indexed: 01/30/2023] Open
Abstract
The upstream stimulatory factors (USFs) are regulators of important cellular processes. Both USF1 and USF2 are supposed to have major roles in metabolism, tissue protection and tumor development. However, the knowledge about the mechanisms that control the function of USFs, in particular in tissue protection and cancer, is limited. Phosphorylation is a versatile tool to regulate protein functions. Thereby, phosphorylation can positively or negatively affect different aspects of transcription factor function including protein stability, protein-protein interaction, cellular localization, or DNA binding. The present review aims to summarize the current knowledge about the regulation of USFs by direct phosphorylation and the consequences for USF functions in tissue protection and cancer.
Collapse
Affiliation(s)
- Tina Horbach
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , Oulu, Finland ; Department of Chemistry, University of Kaiserslautern , Kaiserslautern, Germany
| | - Claudia Götz
- Medical Biochemistry and Molecular Biology, Saarland University , Homburg, Germany
| | - Thomas Kietzmann
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , Oulu, Finland
| | - Elitsa Y Dimova
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , Oulu, Finland
| |
Collapse
|
5
|
Armani F, Andersen ML, Galduróz JCF. Tamoxifen use for the management of mania: a review of current preclinical evidence. Psychopharmacology (Berl) 2014; 231:639-49. [PMID: 24441937 DOI: 10.1007/s00213-013-3397-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 12/04/2013] [Indexed: 12/13/2022]
Abstract
RATIONALE Preliminary data on the efficacy of tamoxifen in reducing manic symptoms of bipolar disorder (BD) suggest that this agent may be a potential treatment for the management of this psychiatric disorder. However, the antimanic properties of tamoxifen have not been fully elucidated, hampering the development and/or use of mood-stabilising drugs that may share its same therapeutic mechanisms of action. Notably, we may gain a greater understanding of the neurobiological and therapeutic properties of tamoxifen by using suitable animal models of mania. OBJECTIVES Here, we review the preclinical studies that have evaluated the effects of tamoxifen to provide an overview of the current progress in our understanding of its antimanic actions, highlighting the critical role of protein kinase C (PKC) as a therapeutic target for the treatment of BD. CONCLUSIONS To date, this field has struggled to make significant progress, and the organisation of an explicit battery of tests is a valuable tool for assessing a number of prominent facets of BD, which may provide a greater understanding of the entire scope of this disease.
Collapse
Affiliation(s)
- Fernanda Armani
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | | | |
Collapse
|
6
|
M T, JE G, RL H, AL H, VB R. The role of PKC signaling in CRF-induced modulation of startle. Psychopharmacology (Berl) 2013; 229:579-89. [PMID: 23722830 PMCID: PMC3784645 DOI: 10.1007/s00213-013-3114-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 04/11/2013] [Indexed: 02/06/2023]
Abstract
RATIONALE Hypersignaling of corticotropin releasing factor (CRF) has been implicated in stress disorders; however, many of its downstream mechanisms of action remain unclear. In vitro, CRF1 receptor activation initiates multiple cell signaling cascades, including protein kinase A (PKA), protein kinase C (PKC), and mitogen-activated protein kinase kinase MEK1/2 signaling. It is unclear, however, which of these signaling cascades mediate CRF-induced behaviors during stress. OBJECTIVES We examined the role of PKA, PKC, and MEK1/2 signaling pathways in CRF-induced anxiety as measured by startle hyperreactivity. METHODS Mice treated with intracerbroventricular (ICV) ovine CRF (oCRF) were pretreated with the PKA inhibitor Rp-cAMPS, PKC inhibitor bisindolylmaleimide (BIM), or MEK1/2 inhibitor PD98059 (ICV) and assessed for acoustic startle reactivity. RESULTS The PKC inhibitor BIM significantly attenuated CRF-induced increases in startle. BIM was also able to block startle increases induced by oCRF when both compounds were infused directly into the bed nucleus of stria terminalis (BNST). PKA and MEK1/2 inhibition had no significant effects on CRF-induced changes in startle at the dose ranges tested. CRF-induced disruption of prepulse inhibition was not significantly reversed by any of the three pretreatments at the dose ranges tested. CONCLUSIONS PKC signaling is required for CRF-induced increases in startle, and this effect is mediated at least in part at the BNST. These findings suggest that PKC signaling cascades (1) may be important for the acute effects of CRF to induce startle hyperreactivity and (2) support further research of the role of PKC signaling in startle abnormalities relevant to disorders such as posttraumatic stress disorder.
Collapse
Affiliation(s)
- Toth M
- Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla CA USA,Dept. of Psychiatry, University of California San Diego, La Jolla CA USA
| | - Gresack JE
- Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla CA USA,Dept. of Psychiatry, University of California San Diego, La Jolla CA USA,Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York NY USA
| | - Hauger RL
- Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla CA USA,Dept. of Psychiatry, University of California San Diego, La Jolla CA USA
| | - Halberstadt AL
- Dept. of Psychiatry, University of California San Diego, La Jolla CA USA
| | - Risbrough VB
- Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla CA USA,Dept. of Psychiatry, University of California San Diego, La Jolla CA USA,Corresponding author: Victoria Risbrough, Ph.D., University of California San Diego, 9500 Gilman Dr. MC0804, La Jolla CA 92093-0804, Tel: 16195433582; Fax: 16195432475:
| |
Collapse
|
7
|
Lidington D, Schubert R, Bolz SS. Capitalizing on diversity: an integrative approach towards the multiplicity of cellular mechanisms underlying myogenic responsiveness. Cardiovasc Res 2012. [PMID: 23180720 DOI: 10.1093/cvr/cvs345] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The intrinsic ability of resistance arteries to respond to transmural pressure is the single most important determinant of their function. Despite an ever-growing catalogue of signalling pathways that underlie the myogenic response, it remains an enigmatic mechanism. The myogenic response's mechanistic diversity has largely been attributed to 'hard-wired' differences across species and vascular beds; however, emerging evidence suggests that the mechanistic basis for the myogenic mechanism is, in fact, 'plastic'. This means that the myogenic response can change quantitatively (i.e. change in magnitude) and qualitatively (i.e. change in mechanistic basis) in response to environmental challenges (e.g. disease conditions). Consequently, understanding the dynamics of how the myogenic response capitalizes on its mechanistic diversity is key to unlocking clinically viable interventions. Using myogenic sphingosine-1-phosphate (S1P) signalling as an example, this review illustrates the remarkable plasticity of the myogenic response. We propose that currently unidentified 'organizational programmes' dictate the contribution of individual signalling pathways to the myogenic response and introduce the concept that certain signalling elements act as 'divergence points' (i.e. as the potential higher level regulatory sites). In the context of pressure-induced S1P signalling, the S1P-generating enzyme sphingosine kinase 1 serves as a divergence point, by orchestrating the calcium-dependent and -independent signalling pathways underlying microvascular myogenic responsiveness. By acting on divergence points, the proposed 'organizational programmes' could form the basis for the flexible recruitment and fine-tuning of separate signalling streams that underlie adaptive changes to the myogenic response and its distinctiveness across species and vascular beds.
Collapse
Affiliation(s)
- Darcy Lidington
- Department of Physiology, University of Toronto, Medical Science Building, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | | | | |
Collapse
|
8
|
Zhao H, Ren C, Chen X, Shen J. From rapid to delayed and remote postconditioning: the evolving concept of ischemic postconditioning in brain ischemia. Curr Drug Targets 2012; 13:173-87. [PMID: 22204317 DOI: 10.2174/138945012799201621] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2010] [Revised: 08/09/2011] [Accepted: 08/12/2011] [Indexed: 01/13/2023]
Abstract
Ischemic postconditioning is a concept originally defined to contrast with that of ischemic preconditioning. While both preconditioning and postconditioning confer a neuroprotective effect on brain ischemia, preconditioning is a sublethal insult performed in advance of brain ischemia, and postconditioning, which conventionally refers to a series of brief occlusions and reperfusions of the blood vessels, is conducted after ischemia/reperfusion. In this article, we first briefly review the history of preconditioning, including the experimentation that initially uncovered its neuroprotective effects and later revealed its underlying mechanisms-of-action. We then discuss how preconditioning research evolved into that of postconditioning--a concept that now represents a broad range of stimuli or triggers, including delayed postconditioning, pharmacological postconditioning, remote postconditioning--and its underlying protective mechanisms involving the Akt, MAPK, PKC and K(ATP) channel cell-signaling pathways. Because the concept of postconditioning is so closely associated with that of preconditioning, and both share some common protective mechanisms, we also discuss whether a combination of preconditioning and postconditioning offers greater protection than preconditioning or postconditioning alone.
Collapse
Affiliation(s)
- Heng Zhao
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305-5327, USA.
| | | | | | | |
Collapse
|
9
|
Effect of variation in diacylglycerol kinase η (DGKH) gene on brain function in a cohort at familial risk of bipolar disorder. Neuropsychopharmacology 2012; 37:919-28. [PMID: 22048461 PMCID: PMC3280657 DOI: 10.1038/npp.2011.272] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Several lines of evidence indicate that the diacylglycerol kinase eta (DGKH) gene is implicated in the etiology of bipolar disorder (BD). However, the functional neural mechanisms of DGKH's risk association remain unknown. Therefore, we examined the effects of three haplotype-tagging risk variants in DGKH (single nucleotide polymorphisms rs9315885, rs1012053, and rs1170191) on brain activation using a verbal fluency functional magnetic resonance imaging task. The subject groups consisted of young individuals at high familial risk of BD (n=81) and a comparison group of healthy controls (n=75). Individuals were grouped based on risk haplotypes described in previous studies. There was a significant risk haplotype*group interaction in the left medial frontal gyrus (BA10, involving anterior cingulate BA32), left precuneus, and right parahippocampal gyrus. All regions demonstrated greater activation during the baseline condition than sentence completion. Individuals at high familial risk for BD homozygous for the DGKH risk haplotype demonstrated relatively greater activation (poor suppression) of these regions during the task vs the low-risk haplotype subjects. The reverse pattern was seen for the control subjects. These findings suggest that there are differential effects of the DGKH gene in healthy controls vs the bipolar high-risk group, which manifests as a failure to disengage default-mode regions in those at familial risk carrying the risk haplotype.
Collapse
|
10
|
Yang P, Li ZQ, Song L, Yin YQ. Protein kinase C regulates neurite outgrowth in spinal cord neurons. Neurosci Bull 2010; 26:117-25. [PMID: 20332816 DOI: 10.1007/s12264-010-1105-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE The functional roles of protein kinase C (PKC) in the neurite outgrowth and nerve regeneration remain controversial. The present study was aimed to investigate the role of PKC in neurite outgrowth, by studying their regulatory effects on neurite elongation in spinal cord neurons in vitro. METHODS The anterior-horn neurons of spinal cord from embryonic day 14 (E14) Sprague-Dawley (SD) rats were dissociated, purified and cultured in the serum-containing medium. The ratio of membrane-PKC (mPKC) activity to cytoplasm-PKC (cPKC) activity (m/c-PKC) was studied at different time points during culture. RESULTS Between 3-11 d of culture, the change of m/c-PKC activity ratio and PKC-betaII expression in the neurite were both significantly correlated with neurite outgrowth (r=0.95, P< 0.01; r=0.73, P< 0.01, respectively). Moreover, PMA, an activator of PKC, induced a dramatic elevation in the m/c-PKC activity ratio, accompanied with the increase in neurite length (r=0.99, P< 0.01). In contrast, GF 109203X, an inhibitor of PKC, significantly inhibited neurite elongation, which could not be reversed by PMA. CONCLUSION PKC activity may be important in regulating neurite outgrowth in spinal cord neurons, and betaII isoform of PKC probably plays a major role in this process.
Collapse
Affiliation(s)
- Ping Yang
- Department of Anatomy, Third Military Medical University, Chongqing 400038, China.
| | | | | | | |
Collapse
|
11
|
Lee HK, Yeo S, Kim JS, Lee JG, Bae YS, Lee C, Baek SH. Protein Kinase C-η and Phospholipase D2 Pathway Regulates Foam Cell Formation via Regulator of G Protein Signaling 2. Mol Pharmacol 2010; 78:478-85. [DOI: 10.1124/mol.110.064394] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
12
|
The response to postnatal stress: amino acids transporters and PKC activity. Neurochem Res 2010; 35:967-75. [PMID: 20306295 DOI: 10.1007/s11064-010-0153-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2010] [Indexed: 10/19/2022]
Abstract
It is well known that animals exposed to stressful stimuli during their early life develop different neurological disorders when they become adults. In this study, we evaluated the effect of acute cold stress on gamma-aminobutyric acid (GABA) and L-Serine (L-Ser) transporters in vitro, using the uptake of [(3)H]-GABA and [(3)H]L-Ser by synaptosomes-enriched fractions isolated from rat cerebral cortex during postnatal development. GABA and L-Ser uptake studies in vitro will be used in this investigation as a colateral evidence of changes in the expression of transporters of GABA and L-Ser. We observed that the maximum velocity (V (max)) in L-Ser and GABA uptake after stress session increased in all stages studied. In contrast, K (m) values of L-Ser uptake enhancent in almost age calculated, excluding at PD21 after cold stress during development, at the same time as K (m) (uptake affinity) values of GABA increased in just about age considered but not at PD5 compared with the control group. Finally we investigated the mechanism by which cells regulate the substrate affinity of L-Ser and GABA transporters. We demonstrated a significantly increase in total PKC activity to PD5 from PD21. Pretreatment with PKC inhibitor: staurosporine (SP) led to a restoration of control uptake in several postnatal-days suggesting a relationship between amino acids system and PKC activation. These findings suggest that a single exposure to postnatal cold stress at different periods after birth modifies both GABA and L-Ser transporters and the related increase in total PKC activity could be intracellular events that participate in neuronal plasticity by early life stress, which could be relevant to function of transporters in the adult rat brain.
Collapse
|
13
|
Machado-Vieira R, Manji HK, Zarate CA. The role of lithium in the treatment of bipolar disorder: convergent evidence for neurotrophic effects as a unifying hypothesis. Bipolar Disord 2009; 11 Suppl 2:92-109. [PMID: 19538689 PMCID: PMC2800957 DOI: 10.1111/j.1399-5618.2009.00714.x] [Citation(s) in RCA: 216] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Lithium has been and continues to be the mainstay of bipolar disorder (BD) pharmacotherapy for acute mood episodes, switch prevention, prophylactic treatment, and suicide prevention. Lithium is also the definitive proof-of-concept agent in BD, although it has recently been studied in other psychoses as well as diverse neurodegenerative disorders. Its neurotrophic effects can be viewed as a unifying model to explain several integrated aspects of the pathophysiology of mood disorders and putative therapeutics for those disorders. Enhancing neuroprotection (which directly involves neurotrophic effects) is a therapeutic strategy intended to slow or halt the progression of neuronal loss, thus producing long-term benefits by favorably influencing outcome and preventing either the onset of disease or clinical decline. The present article: (i) reviews what has been learned regarding lithium's neurotrophic effects since Cade's original studies with this compound; (ii) presents human data supporting the presence of cellular atrophy and death in BD as well as neurotrophic effects associated with lithium in human studies; (iii) describes key direct targets of lithium involved in these neurotrophic effects, including neurotrophins, glycogen synthase kinase 3 (GSK-3), and mitochondrial/endoplasmic reticulum key proteins; and (iv) discusses lithium's neurotrophic effects in models of apoptosis and excitotoxicity as well as its potential neurotrophic effects in models of neurological disorders. Taken together, the evidence reviewed here suggests that lithium's neurotrophic effects in BD are an example of an old molecule acting as a new proof-of-concept agent. Continued work to decipher lithium's molecular actions will likely lead to the development of not only improved therapeutics for BD, but to neurotrophic enhancers that could prove useful in the treatment of many other illnesses.
Collapse
Affiliation(s)
- Rodrigo Machado-Vieira
- Experimental Therapeutics, Mood and Anxiety Disorders Research Program, NIMH-NIH, Department of Health and Human Services, Bethesda, MD
| | - Husseini K Manji
- Johnson and Johnson Pharmaceutical Research and Development, Titusville, NJ, USA
| | - Carlos A Zarate
- Experimental Therapeutics, Mood and Anxiety Disorders Research Program, NIMH-NIH, Department of Health and Human Services, Bethesda, MD
| |
Collapse
|
14
|
Zhao H. Ischemic postconditioning as a novel avenue to protect against brain injury after stroke. J Cereb Blood Flow Metab 2009; 29:873-85. [PMID: 19240739 PMCID: PMC2736291 DOI: 10.1038/jcbfm.2009.13] [Citation(s) in RCA: 156] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ischemic postconditioning initially referred to a stuttering reperfusion performed immediately after reperfusion, for preventing ischemia/reperfusion injury in both myocardial and cerebral infarction. It has evolved into a concept that can be induced by a broad range of stimuli or triggers, and may even be performed as late as 6 h after focal ischemia and 2 days after transient global ischemia. The concept is thought to be derived from ischemic preconditioning or partial/gradual reperfusion, but in fact the first experiment for postconditioning was carried out much earlier than that of preconditioning or partial/gradual reperfusion, in the research on myocardial ischemia. This review first examines the protective effects and parameters of postconditioning in various cerebral ischemic models. Thereafter, it provides insights into the protective mechanisms of postconditioning associated with reperfusion injury and the Akt, mitogen-activated protein kinase (MAPK), protein kinase C (PKC), and ATP-sensitive K+ (K(ATP)) channel cell signaling pathways. Finally, some open issues and future challenges regarding clinical translation of postconditioning are discussed.
Collapse
Affiliation(s)
- Heng Zhao
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California 94305-5327, USA.
| |
Collapse
|
15
|
Zarate CA, Manji HK. Protein kinase C inhibitors: rationale for use and potential in the treatment of bipolar disorder. CNS Drugs 2009; 23:569-82. [PMID: 19552485 PMCID: PMC2802274 DOI: 10.2165/00023210-200923070-00003] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Bipolar disorder is one of the most severely debilitating of all medical illnesses. For a large number of patients, outcomes are quite poor. The illness results in tremendous suffering for patients and their families and commonly impairs functioning and workplace productivity. Risks of increased morbidity and mortality, unfortunately, are frequent occurrences as well. Until recently, little has been known about the specific molecular and cellular underpinnings of bipolar disorder. Such knowledge is crucial for the prospect of developing specific targeted therapies that are more effective and that have a more rapid onset of action than currently available treatments. Exciting recent data suggest that regulation of certain signalling pathways may be involved in the aetiology of bipolar disorder and that these pathways may be profitably targeted to treat the disorder. In particular, mania is associated with overactive protein kinase C (PKC) intracellular signalling, and recent genome-wide association studies of bipolar disorder have implicated an enzyme that reduces the activation of PKC. Importantly, the current mainstays in the treatment of mania, lithium (a monovalent cation) and valproate (a small fatty acid) indirectly inhibit PKC. In addition, recent clinical studies with the relatively selective PKC inhibitor tamoxifen add support to the relevance of the PKC target in bipolar disorder. Overall, a growing body of work both on a preclinical and clinical level indicates that PKC signalling may play an important role in the pathophysiology and treatment of bipolar disorder. The development of CNS-penetrant PKC inhibitors may have considerable benefit for this devastating illness.
Collapse
Affiliation(s)
- Carlos A. Zarate
- Laboratory of Molecular Pathophysiology and Experimental Therapeutics, Bethesda, Maryland, USA, Mood and Anxiety Disorders Research Program, National Institute of Mental Health, Bethesda, Maryland, USA
| | - Husseini K. Manji
- Laboratory of Molecular Pathophysiology and Experimental Therapeutics, Bethesda, Maryland, USA, Mood and Anxiety Disorders Research Program, National Institute of Mental Health, Bethesda, Maryland, USA
| |
Collapse
|
16
|
Zhang H, Forman HJ. Acrolein Induces Heme Oxygenase-1 through PKC-δ and PI3K in Human Bronchial Epithelial Cells. Am J Respir Cell Mol Biol 2008; 38:483-90. [DOI: 10.1165/rcmb.2007-0260oc] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
17
|
Pandey GN, Ren X, Dwivedi Y, Pavuluri MN. Decreased protein kinase C (PKC) in platelets of pediatric bipolar patients: effect of treatment with mood stabilizing drugs. J Psychiatr Res 2008; 42:106-16. [PMID: 17208254 PMCID: PMC2190755 DOI: 10.1016/j.jpsychires.2006.11.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2006] [Revised: 10/31/2006] [Accepted: 11/02/2006] [Indexed: 12/25/2022]
Abstract
Pediatric bipolar disorder (PBD) is a major public health concern, however, its neurobiology is poorly understood. We, therefore, studied the role of protein kinase C (PKC) in the pathophysiology of bipolar illness. We determined PKC activity and immunolabeling of various PKC isozymes (i.e., PKC alpha, PKC betaI, PKC betaII, and PKC delta) in the cytosol and membrane fractions of platelets obtained from PBD patients and normal control subjects. PKC activity and PKC isozymes were also determined after 8 weeks of pharmacotherapy of PBD patients (n=16) with mood stabilizers. PKC activity and the protein expression of PKC betaI and betaII, but not PKC alpha or PKC delta, were significantly decreased in both membrane as well as cytosol fractions of platelets obtained from medication-free PBD patients compared with normal control subjects. Eight weeks of pharmacotherapy resulted in significantly increased PKC activity but no significant changes in any of the PKC isozymes in PBD patients. These results indicate that decreases of specific PKC isozymes and decreased PKC activity may be associated with the pathophysiology of PBD and that pharmacotherapy with mood stabilizing drugs results in an increase and normalization of PKC activity along with improvement in clinical symptoms.
Collapse
Affiliation(s)
- Ghanshyam N Pandey
- University of Illinois at Chicago, Department of Psychiatry, 1601 West Taylor Street, Chicago, IL 60612, USA.
| | | | | | | |
Collapse
|
18
|
Kim JY, Park CS. Potentiation of large-conductance calcium-activated potassium (BK(Ca)) channels by a specific isoform of protein kinase C. Biochem Biophys Res Commun 2007; 365:459-65. [PMID: 17991423 DOI: 10.1016/j.bbrc.2007.10.179] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2007] [Accepted: 10/30/2007] [Indexed: 01/16/2023]
Abstract
The phosphorylation state of large-conductance calcium-activated potassium (BK(Ca)) channels regulates their activity and is dynamically regulated by protein phosphatases and kinases, including protein kinase C (PKC). In this study, we showed that PKC activators up-regulate the activity of the BK(Ca) channel alpha (alpha)-subunit, Slo1, in cell-attached patches of transfected COS7 cells. In an immune complex kinase assay, BK(Ca) channels isolated from rat brain were phosphorylated in the presence of PKC activators, without the addition of exogenous PKC, which suggests that PKC and BK(Ca) channels functionally interact in vivo. Four different PKC isozymes, including PKCdelta, phosphorylated the C-terminus of Slo1 and the addition of purified PKCdelta-activated BK(Ca) channels in excised patches of transfected HEK293 cells. Our results demonstrate that PKC up-regulates BK(Ca) channels and that PKCdelta may functionally interact with BK(Ca) channel complexes in vivo.
Collapse
Affiliation(s)
- Ji-Yoen Kim
- Department of Life Science, Gwangju Institute of Science & Technology, Gwangju 500-712, Republic of Korea
| | | |
Collapse
|
19
|
Wilkie MB, Besheer J, Kelley SP, Kumar S, O'Buckley TK, Morrow AL, Hodge CW. Acute ethanol administration rapidly increases phosphorylation of conventional protein kinase C in specific mammalian brain regions in vivo. Alcohol Clin Exp Res 2007; 31:1259-67. [PMID: 17511744 PMCID: PMC2861774 DOI: 10.1111/j.1530-0277.2007.00423.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Protein kinase C (PKC) is a family of isoenzymes that regulate a variety of functions in the central nervous system including neurotransmitter release, ion channel activity, and cell differentiation. Growing evidence suggests that specific isoforms of PKC influence a variety of behavioral, biochemical, and physiological effects of ethanol in mammals. The purpose of this study was to determine whether acute ethanol exposure alters phosphorylation of conventional PKC isoforms at a threonine 674 (p-cPKC) site in the hydrophobic domain of the kinase, which is required for its catalytic activity. METHODS Male rats were administered a dose range of ethanol (0, 0.5, 1, or 2 g/kg, intragastric) and brain tissue was removed 10 minutes later for evaluation of changes in p-cPKC expression using immunohistochemistry and Western blot methods. RESULTS Immunohistochemical data show that the highest dose of ethanol (2 g/kg) rapidly increases p-cPKC immunoreactivity specifically in the nucleus accumbens (core and shell), lateral septum, and hippocampus (CA3 and dentate gyrus). Western blot analysis further showed that ethanol (2 g/kg) increased p-cPKC expression in the P2 membrane fraction of tissue from the nucleus accumbens and hippocampus. Although p-cPKC was expressed in numerous other brain regions, including the caudate nucleus, amygdala, and cortex, no changes were observed in response to acute ethanol. Total PKCgamma immunoreactivity was surveyed throughout the brain and showed no change following acute ethanol injection. CONCLUSIONS These results suggest that ethanol rapidly promotes phosphorylation of cPKC in limbic brain regions, which may underlie effects of acute ethanol on the nervous system and behavior.
Collapse
Affiliation(s)
- Mary Beth Wilkie
- Bowles Center for Alcohol Studies, Department of Psychiatry, School of Medicine University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Yoshida K. PKCdelta signaling: mechanisms of DNA damage response and apoptosis. Cell Signal 2007; 19:892-901. [PMID: 17336499 DOI: 10.1016/j.cellsig.2007.01.027] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2007] [Revised: 01/19/2007] [Accepted: 01/19/2007] [Indexed: 01/02/2023]
Abstract
The cellular response to genotoxic stress that damages DNA includes cell cycle arrest, activation of DNA repair, and in the event of irreparable damage, induction of apoptosis. However, the signals that determine cell fate, that is, survival or apoptosis, are largely unknown. The delta isoform of protein kinase C (PKCdelta) has been implicated in many important cellular processes, including regulation of apoptotic cell death. The available information supports a model in which certain sensors of DNA lesions activate PKCdelta. This activation is triggered in part by tyrosine phosphorylation of PKCdelta by c-Abl tyrosine kinase. PKCdelta is further proteolytically activated by caspase-3. The cleaved catalytic fragment of PKCdelta translocates to the nucleus and induces apoptosis. Importantly, accumulating data have revealed the nuclear targets for PKCdelta in the induction of apoptosis. A pro-apoptotic function of activated PKCdelta is mediated by at least several downstream effectors known to be associated with the elicitation of apoptosis. Recent findings also demonstrated that PKCdelta is involved in cell cycle-specific activation and induction of apoptotic cell death. Moreover, previous studies have shown that PKCdelta regulates transcription by phosphorylating various transcription factors, including the p53 tumor suppressor that is critical for cell cycle arrest and apoptosis in response to DNA damage. These findings collectively support a pivotal role for PKCdelta in the induction of apoptosis with significant impact. This review is focused on the current views regarding the regulation of cell fate by PKCdelta signaling in response to DNA damage.
Collapse
Affiliation(s)
- Kiyotsugu Yoshida
- Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan.
| |
Collapse
|
21
|
Smith CPS, Oh JD, Bibbiani F, Collins MA, Avila I, Chase TN. Tamoxifen effect on L-DOPA induced response complications in parkinsonian rats and primates. Neuropharmacology 2007; 52:515-26. [PMID: 17116309 DOI: 10.1016/j.neuropharm.2006.08.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2005] [Revised: 06/29/2006] [Accepted: 08/01/2006] [Indexed: 10/23/2022]
Abstract
The contribution of striatal protein kinase C (PKC) isoform changes in levodopa (L-DOPA) induced motor response complications in parkinsonian rats was investigated and the ability of tamoxifen, an antiestrogen with a partial PKC antagonist property, to prevent these response alterations in 6-hydroxydopamine (6-OHDA) lesioned rats as well as in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treated cynomologous monkeys was studied. Following treatment of adult male rats with L-DOPA twice daily for 3 weeks, protein levels of left (lesioned) and right (intact) striatal PKC isoforms were measured. Western blot analysis showed increased protein expression of both the novel PKC epsilon isoform and the atypical PKC lambda isoform ipsilateral to the lesion (174+/-17% for epsilon, 140+/-9% for lambda, of intact striatum in 6-OHDA lesioned plus chronic L-DOPA treated animals) in acute L-DOPA treated rats. No enhancement was observed in PKC immunoreactivity for other isoforms. Tamoxifen (5.0 mg/kg p.o.) significantly attenuated the L-DOPA induced augmentation of protein expression of PKC epsilon and PKC lambda, but had no effect on immunoreactivity for other PKC isoforms. In chronic L-DOPA treated parkinsonian rats, tamoxifen prevented (5.0 mg/kg p.o.) as well as ameliorated (5.0 mg/kg p.o.) the characteristic shortening in duration of motor response to L-DOPA challenge. In MPTP lesioned primates, similar to the ameliorative effect seen in rats, tamoxifen (1 and 3 mg/kg p.o) reduced the appearance of L-DOPA induced dyskinesia by 61% and 55% respectively (p<0.05). These results suggest that changes in specific striatal PKC isoforms contribute to the pathogenesis of L-DOPA induced motor complications and further that drugs able to selectively inhibit these signaling kinases might provide adjunctive benefit in the treatment of Parkinson's disease.
Collapse
MESH Headings
- 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine
- Animals
- Antiparkinson Agents/adverse effects
- Disease Models, Animal
- Drug Administration Schedule
- Drug Interactions
- Dyskinesia, Drug-Induced/drug therapy
- Dyskinesia, Drug-Induced/etiology
- Haplorhini
- Levodopa/adverse effects
- Male
- Models, Biological
- Nerve Tissue Proteins/metabolism
- Oxidopamine
- Parkinson Disease, Secondary/chemically induced
- Parkinson Disease, Secondary/drug therapy
- Protein Kinase C/metabolism
- Rats
- Rats, Sprague-Dawley
- Selective Estrogen Receptor Modulators/therapeutic use
- Tamoxifen/therapeutic use
- Time Factors
Collapse
Affiliation(s)
- C P S Smith
- Experimental Therapeutics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
22
|
Textor B, Sator-Schmitt M, Richter KH, Angel P, Schorpp-Kistner M. c-Jun and JunB Are Essential for Hypoglycemia-MediatedVEGFInduction. Ann N Y Acad Sci 2006; 1091:310-8. [PMID: 17341624 DOI: 10.1196/annals.1378.076] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Physiological conditions like hypoxia or hypoglycemia trigger expression of VEGF, a key regulator of angiogenesis. To elucidate the molecular mechanism underlying the VEGF regulation of hypoglycemia, we investigated the role of AP-1 transcription factor subunits c-Jun and JunB. Using c-jun(-/-) and junB(-/-) mouse embryonic fibroblasts, we demonstrate that both c-Jun and JunB are required for the hypoglycemia-mediated induction of VEGF expression. This process is independent of the master regulator of hypoxic stress HIF-1, as HIF expression and stabilization are not affected by the loss of AP-1 subunits. Analysis of signaling cascades regulating c-Jun and/or JunB activity and/or transcription upon hypoglycemia by application of specific inhibitors of protein kinase C (PKC) or extracellular signal-regulated kinase (ERK) signaling revealed that hypoglycemia-mediated induction of c-Jun is regulated via a PKCalpha-dependent signaling pathway. In contrast, JunB is activated by the MAP kinase ERK for the AP-1 subunits c-Jun and JunB to mediate VEGF regulaltion of hypoglycemia.
Collapse
Affiliation(s)
- Björn Textor
- Division of Signal Transduction and Growth Control, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | | | | | | | | |
Collapse
|
23
|
Shin T, Kim S, Ahn M, Kim H. An immunohistochemical study of protein kinase C in the bovine retina. J Vet Med Sci 2006; 68:71-4. [PMID: 16462121 DOI: 10.1292/jvms.68.71] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The expression of protein kinase C (PKC) was studied in the bovine retina by immunohistochemical analysis. Western blot analysis showed that PKC isoforms, including alpha, betaI, delta and theta, were detected in the bovine retina. By immunohistochemistry, both PKC alpha and betaI were expressed in all retinal layers, with an intense localization of both PKC alpha and betaI detected in bipolar cells in the inner nuclear cell layer and in some glial cells in ganglion cell layers. The immunoreactivity of both PKC delta and theta was quite weak in the retinal layers, compared with that of PKC alpha and betaI. These findings suggest that both conventional and novel PKCs are differentially expressed in the bovine retina.
Collapse
Affiliation(s)
- Taekyun Shin
- Department of Veterinary Medicine, Graduate School, Cheju National University, Jeju city 690-756, Republic of Korea
| | | | | | | |
Collapse
|
24
|
Zhou Y, Wang D, Li F, Shi J, Song J. Different roles of protein kinase C-betaI and -delta in the regulation of adipocyte differentiation. Int J Biochem Cell Biol 2006; 38:2151-63. [PMID: 16950644 DOI: 10.1016/j.biocel.2006.06.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2006] [Revised: 06/08/2006] [Accepted: 06/20/2006] [Indexed: 12/22/2022]
Abstract
Protein kinase C (PKC) is a member of serine/threonine protein kinase family that plays important roles in the control of vast variety of cellular functions. Nevertheless, the regulatory effect of PKC on adipogenesis remained not well understood. In this study, we investigated the effect of several PKC isoforms on the adipogenic conversion of 3T3-L1 preadipocytes induced by dexamethasone, isobutylmethylxanthine and insulin. Treatment of cells with broad-spectrum PKC inhibitor Rö318220 suppressed the adipogenesis. Gö6976, a selective inhibitor for PKC isoforms-alpha, -betaI and -mu, also inhibited the adipogenesis of cells. Pretreatment of cells with peroxisomal proliferator activated receptor-gamma (PPARgamma) agonist troglitazone abolished the inhibitory effect of Gö6976 on adipogenesis. The plasmic membrane translocation of PKC-betaI was observed at the first 2 days of differentiation. Whereas no translocation of PKC-alpha and -mu was observed. Overexpression of dominant negative PKC-betaI, but not wild-type PKC-betaI, blocked adipogenesis. This effect of dominant negative PKC-betaI can be reversed by troglitazone, suggesting that PKC-betaI is required for the initiation of adipogenesis. In addition, rottlerin, a specific inhibitor of PKC-delta, can reverse the suppression of adipogenesis mediated by 12-O-tetradecanoyl-phorbol-13-acetate, transforming growth factor-beta1, and epidermal growth factor. These data suggest that PKC-betaI is important in the induction of adipogenesis, while the PKC-delta has an inhibitory role for adipogenesis.
Collapse
Affiliation(s)
- Yiran Zhou
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | | | | | | | | |
Collapse
|
25
|
Toroser D, Yarian CS, Orr WC, Sohal RS. Mechanisms of gamma-glutamylcysteine ligase regulation. BIOCHIMICA ET BIOPHYSICA ACTA 2006; 1760:233-44. [PMID: 16324789 PMCID: PMC2837077 DOI: 10.1016/j.bbagen.2005.10.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2005] [Revised: 10/17/2005] [Accepted: 10/23/2005] [Indexed: 01/21/2023]
Abstract
The principal objective of this study was to investigate the mechanisms regulating the activity of gamma-glutamylcysteine ligase (GCL; EC 6.3.2.2), the rate limiting enzyme in glutathione biosynthesis. Two phylogenetically divergent species, mouse and the fruitfly, Drosophila melanogaster were used to test the hypothesis that reversible protein phosphorylation and pyridine dinucleotide phosphate dependent allostery regulate GCL activity. GCL was almost completely inhibited under phosphorylating conditions, involving preincubations with MgATP and endogenous protein kinases. Maximal GCL inhibitions of 94%, 77%, 85%, 87%, 83%, 95% and 89% occurred, respectively, in mouse cerebellum, hippocampus, brainstem, striatum, cortex and heart, and Drosophila. These changes in GCL activity were detected using saturating levels of substrates, suggesting that V(max) was dramatically affected, whereas K(m) values showed no differences. In vitro activation of GCL, presumably due to dephosphorylation, was blocked by inhibitors of protein phosphatases, suggesting that GCL exists in vivo as a mixture of phosphorylated and dephosphorylated forms. The reversibility of the dephosphorylation-dependent activation was indicated by the time-dependent inactivation of the in vitro activated Drosophila GCL, by preincubation with MgATP. NADPH increased maximal GCL activity by up to 93%, whereas several other nucleotide analogues did not, thereby demonstrating specificity. Kinetic analysis using Hanes-Woolf replots of initial velocity data suggested that the NADPH-dependent stimulation of GCL activity is brought about by a change in the maximal activity, V(max), rather than changes in substrate affinity. Results of this study suggest that mechanisms of modulation of eukaryotic GCL enzymes may include specific binding of ligands such as pyridine dinucleotide phosphates and reversible protein phosphorylation.
Collapse
Affiliation(s)
- Dikran Toroser
- Department of Molecular Pharmacology and Toxicology, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089-9121, USA
| | - Connie S. Yarian
- Department of Molecular Pharmacology and Toxicology, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089-9121, USA
| | - William C. Orr
- Department of Biological Sciences, Dedman Life Sciences Building, Southern Methodist University, Dallas, TX 75275, USA
| | - Rajindar S. Sohal
- Department of Molecular Pharmacology and Toxicology, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089-9121, USA
| |
Collapse
|
26
|
Pham TNQ, Brown BL, Dobson PRM, Richardson VJ. Protein kinase C-eta (PKC-eta) is required for the development of inducible nitric oxide synthase (iNOS) positive phenotype in human monocytic cells. Nitric Oxide 2004; 9:123-34. [PMID: 14732335 DOI: 10.1016/j.niox.2003.09.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Several murine and human monocytic cell lines and monocyte-derived macrophages (MDM) from healthy volunteers were studied to compare their production of nitric oxide (NO) and induction of iNOS following endotoxin treatment. Although the human cells were sensitive to endotoxin and responded well by producing TNF-alpha and matrix metalloproteases (MMP), there was no induction of iNOS expression or NO production by any of these cells. Murine cells, however, produced large amounts of NO and expressed iNOS following similar endotoxin stimulation. We investigated the expression of PKC isotypes in all human and murine cell lines as well as in MDM, and found that the human cells lacked PKC-eta while the murine counterparts lacked PKC-beta1. Subsequently, human cells that were transfected with PKC-eta were found to make large quantities of NO following endotoxin exposure, an observation not seen in untransfected cells. We propose that PKC-eta is essential for the development of the iNOS positive phenotype in human monocytic cells, and may be responsible for the development of a number of inflammatory related conditions. As such it may be a suitable target for therapeutic intervention.
Collapse
Affiliation(s)
- Tram N Q Pham
- Basic Medical Sciences, Faculty of Medicine, Memorial University, St. John's NL, Canada A1B 3V6
| | | | | | | |
Collapse
|
27
|
Gentile MT, Vecchione C, Maffei A, Aretini A, Marino G, Poulet R, Capobianco L, Selvetella G, Lembo G. Mechanisms of soluble beta-amyloid impairment of endothelial function. J Biol Chem 2004; 279:48135-42. [PMID: 15319431 DOI: 10.1074/jbc.m407358200] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Alzheimer's disease (AD) has been recently associated with vascular risk factors. beta-amyloid peptides (AbetaP), the main component of senile plaques typical of AD, circulate in soluble globular form in bloodstream. Interestingly, AbetaP is able to induce endothelial dysfunction, and this effect may represent the link between vascular and neuronal pathophysiological factors involved in AD. We aimed to clarify the molecular mechanisms underlying globular AbetaP-induced vascular toxicity. Using several methodological approaches, we have observed that in vascular tissues globular AbetaP is unable to induce oxidative stress, one of the mechanisms hypothesized involved in beta-amyloid toxicity. More important, we have demonstrated that globular AbetaP is able to localize on vascular endothelium, where it inhibits eNOS enzymatic activity. In particular, AbetaP enhances eNOS phosphorylation on threonine 495 and serine 116 and reduces acetylcholine-induced phosphorylation on serine 1177. Such an effect depends on a PKC signaling pathway, as suggested by its phosphorylation on serine 660. In fact, selective inhibition of the calcium-dependent group of PKC is able to rescue beta-amyloid-induced alteration of eNOS phosphorylation, NO production, and endothelial vasorelaxation. The activation of these Ca(2+)-dependent pathways is probably due to the ability of AbetaP to evoke Ca(2+) leakage from inositol 1,4,5-triphosphate receptors on endoplasmic reticulum. Our data demonstrate that globular AbetaP-induced endothelial NO dysfunction can be attributed to an alteration of intracellular Ca(2+) homeostasis, which could lead to the activation of calcium-dependent group of PKC with a consequent change of the eNOS phosphorylation pattern. These mechanisms could contribute to shed further light on the toxic effect of beta-amyloid in vascular tissues.
Collapse
|
28
|
Beom S, Cheong D, Torres G, Caron MG, Kim KM. Comparative Studies of Molecular Mechanisms of Dopamine D2 and D3 Receptors for the Activation of Extracellular Signal-regulated Kinase. J Biol Chem 2004; 279:28304-14. [PMID: 15102843 DOI: 10.1074/jbc.m403899200] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Dopamine D(2) and D(3) receptors (D(2)R/D(3)R), which have similar structural architecture as well as functional similarities, are expressed in the same brain dopaminergic neurons. It is intriguing that two receptor proteins with virtually the same functional roles are expressed in the same neuron. Recently we have shown that D(2)R and D(3)R possess different regulatory processes including intracellular trafficking properties, which implies that they might employ different signaling mechanisms for regulation of the same cellular processes. Here we studied the signaling pathways of ERK activation mediated by D(2)R and D(3)R in HEK-293 cells and corroborated them with concomitant studies in COS-7 cells and C6 cells. Our results show that Src, phosphatidylinositol 3-kinase, and atypical protein kinase C were commonly involved in D(2)R-/D(3)R-mediated ERK activation. However, beta-arrestin and sequestration of D(2)R/D(3)R were found not to be involved. ERK activations mediated by D(3)R, but not D(2)R, were blocked by betaARK-CT, AG1478 epidermal growth factor receptor (EGFR) inhibitor, and by dominant negative mutants of Ras and Raf, suggesting the involvement of the Gbetagamma(i) pathway. The alpha-subunit of G(o) (Galpha(o)) was able to couple with D(3)R to mediate ERK activation. We conclude that D(3)R mainly utilizes the betagamma pathway of G(i) protein, which involves the transactivation of EGFR in HEK-293 cells. In contrast, the alpha-subunit of the G(i) protein plays a main role in D(2)R-mediated ERK activation. Our study suggests one example of intricate cellular regulations in the brain, that is, dopaminergic neurons could regulate ERK activity more flexibly through alternative usage of either the D(2)R or D(3)R pathway depending on the cellular situation.
Collapse
Affiliation(s)
- SunRyeo Beom
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Kwang-Ju, 500-757 Korea
| | | | | | | | | |
Collapse
|
29
|
Jayanthi LD, Samuvel DJ, Ramamoorthy S. Regulated Internalization and Phosphorylation of the Native Norepinephrine Transporter in Response to Phorbol Esters. J Biol Chem 2004; 279:19315-26. [PMID: 14976208 DOI: 10.1074/jbc.m311172200] [Citation(s) in RCA: 122] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The effects of norepinephrine in the brain and periphery are terminated primarily by active reuptake of the catecholamine via cocaine- and amphetamine-sensitive norepinephrine transporters (NETs). Activation of protein kinase C (PKC) down-regulates NET by sequestering it from the plasma membrane, although the underlying mechanism is not yet known. Previously, we showed robust expression of endogenous NETs in rat placental trophoblasts (Jayanthi, L. D., Vargas, G., and DeFelice, L. J. (2002) Br. J. Pharmacol. 135, 1927-1934). Here we report a significant reduction in native NET function and surface expression in these cells following phorbol ester (beta-PMA) treatment. The beta-PMA-mediated down-regulation of NET occurs by a rapid sequestration of NETs from the plasma membrane and is calcium-independent. Reversible biotinylation experiments revealed a significant enhancement of NET endocytosis following beta-PMA treatment. Chemical treatments and expression of dominant negative mutants of dynamin 1 and 2 failed to prevent the beta-PMA effect, suggesting a clathrin-independent pathway. In contrast, treatment with the cholesterol-disrupting agent filipin, which blocks caveolae/lipid raft-mediated internalization, completely blocked the beta-PMA-mediated NET sequestration. Discontinuous sucrose density gradient centrifugation revealed NET in the lipid raft fractions. Following beta-PMA treatment, there was reduced NET levels in the lipid raft fractions suggesting that cholesterol-rich lipid rafts mediate PKC-triggered NET internalization. Metabolic labeling and immunoprecipitation studies revealed that NET phosphorylation is stimulated severalfold by PKC activation and protein phosphatase 1/2A inhibition. Together, these findings demonstrate for the first time that in trophoblasts (i) PKC activation regulates NET function and surface expression by an enhanced internalization process that is lipid raft-mediated and (ii) PKC and protein phosphatase(s) modulation regulates NET phosphorylation.
Collapse
Affiliation(s)
- Lankupalle D Jayanthi
- Department of Physiology and Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425, USA.
| | | | | |
Collapse
|
30
|
Choe Y, Jung H, Khang I, Kim K. Selective roles of protein kinase C isoforms on cell motility of GT1 immortalized hypothalamic neurones. J Neuroendocrinol 2003; 15:508-15. [PMID: 12694376 DOI: 10.1046/j.1365-2826.2003.01023.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Recently, we demonstrated that activation of the protein kinase C (PKC) signalling pathway promoted morphological differentiation of GT1 hypothalamic neurones via an increase in beta-catenin, a cell-cell adhesion molecule, indicating a possible involvement of PKC in cellular motility. In this study, we explored the differential roles of PKC isoforms in GT1 cell migration. First, we transiently transfected GT1 cells with enhanced green fluorescence protein (EGFP)-tagged actin to monitor the dynamic rearrangement of filamentous-actin (F-actin) in living cells. Treatment with 12-O-tetradecanoylphorbol-13-acetate (TPA), a PKC activator, markedly promoted lamellipodia formation, while safingol (a PKC alpha-selective inhibitor) blocked the TPA-induced lamellipodial actin structure. Both wound-healing and Boyden migration assays showed that TPA treatment promoted neuronal migration of GT1 cells; however, cotreatment of TPA with safingol or rottlerin (a PKC delta-selective inhibitor) clearly blocked this TPA effect, indicating that both PKC alpha and PKC delta may be positive regulators of neuronal migration. By contrast, PKC gamma-EGFP-expressing GT1 cells exhibited decreased cellular motility and weak staining for actin stress fibres, suggesting that PKC gamma may act as a negative mediator of cell migration in these neurones. Among the PKC downstream signal molecules, p130Cas, a mediator of cell migration, and its kinase, focal adhesion kinase (FAK), increased following TPA treatment; phosphorylation of p130Cas was induced in a PKC alpha-dependent manner. Together, these results demonstrate that PKC alpha promotes GT1 neuronal migration by activating focal adhesion complex proteins such as p130Cas and FAK.
Collapse
Affiliation(s)
- Y Choe
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | | | | | | |
Collapse
|
31
|
Villa I, Dal Fiume C, Maestroni A, Rubinacci A, Ravasi F, Guidobono F. Human osteoblast-like cell proliferation induced by calcitonin-related peptides involves PKC activity. Am J Physiol Endocrinol Metab 2003; 284:E627-33. [PMID: 12556355 DOI: 10.1152/ajpendo.00307.2002] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The calcitonin peptides [calcitonin (CT), calcitonin gene-related peptide (CGRP), amylin] share many biological actions, including activity on bone cells. In the present study, CT (10(-11) to 10(-9) M) stimulated [(3)H]thymidine incorporation in primary cultures of human osteoblasts (hOB), as already demonstrated for CGRP and amylin. RT-PCR analysis showed that the calcitonin receptor and the calcitonin receptor-like receptor are both expressed in hOB. In these cells, CT (10(-10) M) and amylin (10(-9) M), in contrast to CGRP (10(-8) M), did not increase cAMP production. All three peptides stimulated protein kinase C (PKC) activity. To evaluate PKC involvement in hOB proliferation, cells were incubated with phorbol 12,13-dibutyrate, a stimulator of PKC activity; cell proliferation was increased in a dose-dependent manner (EC(50) = 3.4 x 10(-8) M). Staurosporine (10(-9) M), a PKC inhibitor, blocked phorbol 12,13-dibutyrate-induced PKC activity and cell proliferation. Inhibition of PKC by staurosporine also counteracted the stimulatory effect of CT, CGRP, and amylin on hOB proliferation. From these data, it is deduced that the activation of PKC is important for hOB proliferation and that it is involved in the anabolic effect of CT peptides on bone.
Collapse
Affiliation(s)
- I Villa
- Bone Metabolic Unit, Scientific Institute H San Raffaele, 20132 Milan, Italy
| | | | | | | | | | | |
Collapse
|
32
|
Shahak H, Slotkin TA, Yanai J. Alterations in PKCgamma in the mouse hippocampus after prenatal exposure to heroin: a link from cell signaling to behavioral outcome. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2003; 140:117-25. [PMID: 12524182 DOI: 10.1016/s0165-3806(02)00607-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Administration of heroin to pregnant mice evokes neurochemical and behavioral deficits consequent to disruption of septohippocampal cholinergic innervation, notably involving desensitization of the ability of cholinergic receptors to activate PKC activity. The present study further evaluates whether desensitization occurs specifically for the PKCgamma isoform, the behaviorally relevant subtype, as compared to PKCalpha. Mice were exposed transplacentally to heroin on gestational days (GD) 9-18 via s.c. maternal injections (10 mg/kg per day). In young adulthood (50 days old), control offspring showed an increase in hippocampal cell membrane PKCgamma after incubation with the muscarinic cholinergic receptor agonist, carbachol, indicative of translocation from the cytosol. Prenatal exposure to heroin eliminated this response, whereas basal PKCgamma levels were unchanged. In contrast, PKCalpha, which is not related to heroin-induced behavioral deficits, did not show a loss of response. The present findings strongly point to abnormalities in the responsiveness of PKCgamma as a mechanism underlying the neurobehavioral teratogenicity of heroin.
Collapse
Affiliation(s)
- Halit Shahak
- The Ross Laboratory for Studies in Neural Birth Defects, Department of Anatomy and Cell Biology, The Hebrew University-Hadassah Medical School, P.O. Box 12272, 91120 Jerusalem, Israel
| | | | | |
Collapse
|
33
|
Selvatici R, Marino S, Piubello C, Rodi D, Beani L, Gandini E, Siniscalchi A. Protein kinase C activity, translocation, and selective isoform subcellular redistribution in the rat cerebral cortex after in vitro ischemia. J Neurosci Res 2003; 71:64-71. [PMID: 12478614 DOI: 10.1002/jnr.10464] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Protein kinase C (PKC) involvement in ischemia-induced neuronal damage has been investigated in superfused rat cerebral cortex slices submitted to 15 min of oxygen-glucose deprivation (OGD) and in primary cultures of rat cortical neurons exposed to 100 microM glutamate (GLU) for 10 min. OGD significantly increased the total PKC activity in the slices, mostly translocated in the particulate fraction. After 1 hr of reperfusion, the total PKC activity was reduced and the translocated fraction dropped by 84% with respect to the control. Western blot analysis of OGD samples showed an increase in total beta(2) and epsilon PKC isoform levels. After reperfusion, the total levels of alpha, beta(1), beta(2) and gamma isoforms were significantly reduced, whereas the epsilon isoform remained at an increased level. Endogenous GLU release from OGD slices increased to about 15 times the basal values after 15 min of oxygen-glucose deprivation, and to 25 and 35 times the basal level in the presence of the PKC inhibitors staurosporine (0.1 microM) and bisindolylmaleimide (1 microM), respectively. Western blot analysis of GLU-treated cortical neurons showed a significant decrease only in the total level of beta(2) isoforms. Cell survival was reduced to 31% in GLU-treated neuronal cultures; PKC inhibitors were not able to modify this effect. These findings demonstrate that the cell response to OGD and GLU involves PKC in a complex way. The net role played by PKC during OGD may be to reduce GLU release and, consequently, neurotoxicity. The isoforms beta(2) and epsilon are affected the most and may play a significant role in the mechanisms underlying neurotoxicity/neuroprotection.
Collapse
Affiliation(s)
- Rita Selvatici
- Department of Experimental and Diagnostic Medicine, Section of Medical Genetics, University of Ferrara, Italy.
| | | | | | | | | | | | | |
Collapse
|
34
|
Caputto BL, Guido ME. Shedding light on the metabolism of phospholipids in the retina. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1583:1-12. [PMID: 12069844 DOI: 10.1016/s1388-1981(02)00209-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Beatriz L Caputto
- CIQUIBIC-Departamento de Química Biológica, Facultad de Ciencias Químicas-Universidad Nacional de Córdoba, Pabellón Argentina-Ciudad Universitaria, Argentina.
| | | |
Collapse
|
35
|
Salli U, Stormshak F. Prostaglandin F2alpha-activated protein kinase Calpha phosphorylates myristoylated alanine-rich C kinase substrate protein in bovine luteal cells. Endocrine 2001; 16:83-8. [PMID: 11887938 DOI: 10.1385/endo:16:2:083] [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: 11/11/2022]
Abstract
Prostaglandin F2alpha (PGF2alpha)-induced secretion of oxytocin by the bovine corpus luteum involves the phosphorylation of a unique protein kinase C (PKC) substrate, myristoylated alanine-rich C kinase substrate (MARCKS) protein. This study was conducted to determine the specific PKC isoform engaged in phosphorylation of MARCKS protein in bovine luteal cells. In experiment 1, dispersed luteal cells recovered from the corpus luteum on d 8 of the estrous cycle were preincubated with [32P] orthophosphate and then exposed to PGF2alpha alone or in combination with PKC inhibitors. Autoradiography and densitometry of Western blots revealed that MARCKS protein was phosphorylated by a conventional PKC (cPKC) isoform. Experiment 2 was conducted to identify the specific cPKC isoform that phosphorylates MARCKS protein in luteal cells. Corpora lutea were removed from control and PGF2alpha-treated heifers on d 8 of the cycle, and PKC isoforms associated with membrane and cytosolic fractions were determined. Treatment with PGF2alpha increased membrane concentrations of PKCalpha within 5 min after treatment (p < 0.005). Collectively, these data suggest that phosphorylation of MARCKS protein coinciding with oxytocin secretion is mediated by PKCalpha.
Collapse
Affiliation(s)
- U Salli
- Department of Biochemistry/Biophysics, Oregon State University, Corvallis 97331, USA
| | | |
Collapse
|
36
|
Affiliation(s)
- L Cartee
- Department of Hematology/Oncology, Medical College of Virginia, Richmond, USA
| | | |
Collapse
|
37
|
Mathur A, Vallano ML. 2,2',3,3',4,4'-Hexahydroxy-1,1'-biphenyl-6,6'-dimethanol dimethyl ether (HBDDE)-induced neuronal apoptosis independent of classical protein kinase C alpha or gamma inhibition. Biochem Pharmacol 2000; 60:809-15. [PMID: 10930535 DOI: 10.1016/s0006-2952(00)00398-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Protein kinase C (PKC) isozymes constitute a family of at least 12 structurally related serine-threonine kinases that are differentially regulated and localized, and are presumed to mediate distinct intracellular functions. To explore their roles in intact cells, investigators are developing cell-permeable, isoform-selective inhibitors. 2,2',3,3',4,4'-Hexahydroxy-1, 1'-biphenyl-6,6'-dimethanol dimethyl ether (HBDDE) is reported to be a selective inhibitor of PKC alpha and gamma with IC(50) values of 43 and 50 microM, respectively, using an in vitro assay. However, data examining the potency and selectivity of HBDDE in intact cells are lacking. Employing rodent cerebellar granule neurons as a model system, we investigated the effects of HBDDE using cell survival as a functional end-point. HBDDE induced an apoptotic form of cell death that was dependent upon protein synthesis and included activation of a terminal executioner of apoptosis, caspase 3. The concentration of HBDDE required for half-maximal cell death was less than 10 microM ( approximately 5-fold less than the reported IC(50) values for PKC alpha and gamma in vitro). Furthermore, HBDDE induced apoptosis even after phorbol-ester-mediated down-regulation of PKC alpha and gamma, indicating that this effect is independent of these isoforms. Consistent with this, 2-[1-(3-dimethylaminopropyl) indol-3-yl]-3-(indol-3-yl)-maleimide (GF 109203X), a general inhibitor of all classical and some novel PKCs, did not interfere with survival. Thus, HBDDE should not be used as an isoform-selective inhibitor of PKC alpha or gamma in intact cells. Nevertheless, identification of its target in granule neurons will provide valuable information about survival pathways.
Collapse
Affiliation(s)
- A Mathur
- Department of Pharmacology, Upstate Medical University, Syracuse, NY 13210, USA
| | | |
Collapse
|
38
|
Huang Z, Ghalayini A, Guo XX, Alvarez KM, Anderson RE. Light-mediated activation of diacylglycerol kinase in rat and bovine rod outer segments. J Neurochem 2000; 75:355-62. [PMID: 10854281 DOI: 10.1046/j.1471-4159.2000.0750355.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The hydrolysis of phosphatidylinositol 4,5-bisphosphate is regulated by light in retinal rod outer segment (ROS) membranes. We recently reported that the activities of phosphatidylinositol synthetase and phosphatidylinositol 3-kinase are also higher in bleached (light-exposed) ROS (B-ROS). In this study, we investigated the effect of bleaching on diacylglycerol (DAG) kinase (DAG-kinase) activity in bovine and rat ROS membranes prepared from dark-adapted (D-ROS) or bleached (B-ROS) retinas. In bovine ROS, DAG-kinase activity toward endogenous DAG substrate was higher in B-ROS than in D-ROS. Quantification of DAG in both sets of membranes showed that the levels were the same, eliminating the possibility that the greater DAG-kinase activity was due to higher levels of endogenous substrate in B-ROS. DAG-kinase activity was also higher in B-ROS against an exogenous, water-soluable substrate (1, 2-didecanoyl-rac-glycerol), which competed with endogenous DAG substrate and saturated at approximately 2 mM. Immunoblot analysis with an anti-DAG-kinase gamma polyclonal antibody demonstrated that the gamma isoform was present in isolated bovine ROS. Immunocytochemistry of frozen bovine retinal sections confirmed the presence of DAG-kinase gamma immunoreactivity in ROS, as well as other retinal cells. Quantification of the immunoreactive products on western blots showed that more DAG-kinase gamma was present in B-ROS than in D-ROS. In an in vivo experiment, ROS prepared from rats exposed to 30 min of room light had greater DAG-kinase activity than ROS prepared from dark-adapted animals. Taken together, these data suggest that light exposure leads to the translocation of DAG-kinase from the cytosol to ROS membranes and that the greater DAG-kinase activity in B-ROS is due to the presence of more protein associated with ROS membranes.
Collapse
Affiliation(s)
- Z Huang
- Departments of Ophthalmology, Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center Dean A. McGee Eye Institute, Oklahoma City, Oklahoma, USA
| | | | | | | | | |
Collapse
|
39
|
Clemow DB, Steers WD, Tuttle JB. Stretch-activated signaling of nerve growth factor secretion in bladder and vascular smooth muscle cells from hypertensive and hyperactive rats. J Cell Physiol 2000; 183:289-300. [PMID: 10797303 DOI: 10.1002/(sici)1097-4652(200006)183:3<289::aid-jcp1>3.0.co;2-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Elevated vascular (VSMC) and bladder smooth muscle (BSMC) NGF are associated with altered visceral innervation in the spontaneously hypertensive rat (SHR: hypertensive, behaviorally hyperactive) compared with control Wistar-Kyotos (WKYs). Stretch stimulates increased NGF production in BSMCs. To elucidate whether stretch induces NGF synthesis in VSMCs, and to determine if disturbances in stretch-mediated NGF production contribute to the elevated tissue levels of NGF in SHRs, we subjected VSMCs and BSMCs cultured from four established inbred rat strains (WKY, WKHA: hyperactive; SHR and WKHT: hypertensive) to several stretch paradigms. For VSMCs, acute and cyclic stretch affected cells derived from hypertensive rats (80-100% increase over control) but not from normotensive strains. For BSMCs, cyclic and static stretch increased NGF secretion in all four strains, but had a two- to threefold greater effect in cells from SHRs and WKHTs (increase up to 600%) at early time points. At later time points of a 24-h experimental period, stretch increased NGF output up to 400% in SHR and WKHA cultures. Thus, defects that influence early induction of stretch-mediated SHR NGF secretion cosegregate with the hypertensive phenotype. Stretch-gated ion channel inhibitors, voltage-gated ion channel inhibitors, and protease inhibitors failed to affect stretch-induced BSMC NGF secretion. In contrast, gene transcription, intracellular calcium, protein kinase C (PKC), and autocrine release of an unknown factor may play a role in the elevated NGF secretion observed in smooth muscle from hypertensive animals. Altered stretch-induced smooth muscle NGF secretion may contribute to the augmented vascular and bladder NGF content associated with high blood pressure and hyperactive voiding in SHRs.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/physiology
- Aorta, Thoracic/physiopathology
- Attention Deficit Disorder with Hyperactivity/physiopathology
- Cells, Cultured
- Hypertension/physiopathology
- Muscle, Smooth/physiology
- Muscle, Smooth/physiopathology
- Muscle, Smooth, Vascular/physiology
- Muscle, Smooth, Vascular/physiopathology
- Nerve Growth Factors/genetics
- Nerve Growth Factors/metabolism
- Rats
- Rats, Inbred SHR
- Rats, Inbred WKY
- Rats, Sprague-Dawley
- Rats, Wistar
- Signal Transduction
- Stress, Mechanical
- Transcription, Genetic
- Urinary Bladder/physiology
- Urinary Bladder/physiopathology
Collapse
Affiliation(s)
- D B Clemow
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA
| | | | | |
Collapse
|
40
|
Jayanthi LD, Wilson JJ, Montalvo J, DeFelice LJ. Differential regulation of mammalian brain-specific proline transporter by calcium and calcium-dependent protein kinases. Br J Pharmacol 2000; 129:465-70. [PMID: 10711344 PMCID: PMC1571857 DOI: 10.1038/sj.bjp.0703071] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. This study examined the role of [Ca2+]I and Ca(2+)-dependent kinases in the modulation of high-affinity, mammalian brain-specific L-proline transporter (PROT). 2. beta-PMA (phorbol 12-myristate 13-acetate), an activator of protein kinase C (PKC), inhibits PRO uptake, and bisindolymalemide I (BIM), a potent PKC inhibitor, prevents beta-PMA inhibition. Down-regulation of PKC by chronic treatment with beta-PMA enhances PROT function indicating PROT regulation by tonic activity of PKC. 3. Thapsigargin, which increases [Ca2+]I levels by inhibiting Ca(2+)-ATPase, inhibits PROT and exhibits additive inhibition when co-treated with beta-PMA. KN-62, a Ca2+/calmodulin-dependent kinase II (CaMK II) inhibitor, but not BIM (a PKC inhibitor) prevents the inhibition by thapsigargin. These data suggest that PKC and CaMK II modulate PROT and that thapsigargin mediates its effect via CaMK II. 4. Thapsigargin raises [Ca2+]I and increases PRO-induced current on a second time scale, whereas the inhibitory effect of thapsigargin occurs only after 10 min of treatment. These data suggest that Ca2+ differentially regulate PROT: Ca2+ initially enhances PRO transport but eventually inhibits transport function through CaMK II pathway. 5. Ca(2+)-induced stimulation exemplifies the acute regulation of a neurotransmitter transporter, which may play a critical role in the profile of neurotransmitters during synaptic transmission.
Collapse
Affiliation(s)
- L D Jayanthi
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-660, USA
| | | | | | | |
Collapse
|
41
|
Pandey GN, Dwivedi Y, Pandey SC, Teas SS, Conley RR, Roberts RC, Tamminga CA. Low phosphoinositide-specific phospholipase C activity and expression of phospholipase C beta1 protein in the prefrontal cortex of teenage suicide subjects. Am J Psychiatry 1999; 156:1895-901. [PMID: 10588402 DOI: 10.1176/ajp.156.12.1895] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The enzyme phosphoinositide-specific phospholipase C (PI-PLC) is a component of the phosphoinositide signal transduction system. Other components of this system have been found to be abnormal in adults and adolescents who have committed suicide, and so the authors examined whether PI-PLC activity and protein expression of PLC isozymes are abnormal in postmortem brains of teenage suicide subjects. METHOD PI-PLC activity and protein expression of the PLC beta1, delta1, and gamma1 isozymes were examined in Brodmann's areas 8 and 9 of postmortem brains obtained from 18 teenage suicide subjects and 18 matched comparison subjects. PI-PLC activity was determined by enzymatic assay, and protein expression of the PLC isozymes was determined by the Western blot technique. RESULTS Compared with the normal subjects, the teenage suicide subjects had significantly lower PI-PLC activity and immunolabeling of the specific PLC beta1 isozyme in both membrane and cytosol fractions of Brodmann's areas 8 and 9 combined (prefrontal cortex). There was also a significant correlation between PI-PLC activity and protein levels of the PLC beta1 isozyme in the brains of the teenage suicide subjects. There was no significant difference in PI-PLC activity or level of PLC beta1 protein between the suicide subjects with a history of mental disorders and those with no history of mental disorders; however, both groups had significantly lower PI-PLC activity and expression of PLC beta1 protein than the normal subjects. CONCLUSIONS Low PI-PLC activity and expressed levels of the PLC beta1 isozyme in postmortem brains of suicide subjects may have clinical relevance in the pathophysiology of suicidal behavior.
Collapse
Affiliation(s)
- G N Pandey
- Psychiatric Institute, Department of Psychiatry, University of Illinois at Chicago, 60612, USA.
| | | | | | | | | | | | | |
Collapse
|
42
|
Cussac D, Newman-Tancredi A, Pasteau V, Millan MJ. Human dopamine D(3) receptors mediate mitogen-activated protein kinase activation via a phosphatidylinositol 3-kinase and an atypical protein kinase C-dependent mechanism. Mol Pharmacol 1999; 56:1025-30. [PMID: 10531409 DOI: 10.1124/mol.56.5.1025] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The mitogen-activated protein kinase (MAPK) cascade is stimulated by both receptor tyrosine kinases and G protein-coupled receptors. We show that recombinant human dopamine D(3) receptors expressed in Chinese hamster ovary cells transiently activate MAPK via pertussis toxin-sensitive Gi and/or Go proteins. The involvement of D(3) receptors was confirmed by use of the D(3) agonists PD 128,907 and (+)-7-hydroxy-2-dipropylaminotetralin, which mimicked the response to dopamine (DA). Furthermore, haloperidol and the selective D(3) receptor antagonists S 14297 and GR 218,231 attenuated DA-induced MAPK activation; however, when tested alone, S 14297 weakly stimulated MAPK activity, suggesting partial agonist activity. The transduction mechanisms by which hD(3) receptors activate MAPK were explored with specific kinase inhibitors. Genistein and lavendustin A, inhibitors of tyrosine kinase activity, did not reduce DA-induced MAPK activation. In contrast, PD 98059, an inhibitor of MAPK kinase, and Ro 31-8220 and Gö 6983, inhibitors of protein kinase C (PKC), blocked DA-induced MAPK activation. However, MAPK activation was insensitive to PKC down-regulation by phorbol esters, indicating the involvement of an "atypical" PKC. Furthermore, MAPK activation involved phosphatidylinositol 3-kinase inasmuch as its inhibition by LY 294002 and wortmannin reduced DA-induced MAPK activation. In conclusion, this study demonstrates that stimulation of hD(3) receptors activates MAPK. This action is mediated via an atypical isoform of PKC, possibly involving cross-talk with products of phosphatidylinositol 3-kinase activation.
Collapse
Affiliation(s)
- D Cussac
- Department of Psychopharmacology, Institut de Recherches Servier, Croissy (Paris), France
| | | | | | | |
Collapse
|
43
|
Shih SC, Mullen A, Abrams K, Mukhopadhyay D, Claffey KP. Role of protein kinase C isoforms in phorbol ester-induced vascular endothelial growth factor expression in human glioblastoma cells. J Biol Chem 1999; 274:15407-14. [PMID: 10336429 DOI: 10.1074/jbc.274.22.15407] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aberrant expression of the potent angiogenic cytokine, vascular endothelial growth factor (VEGF), has been demonstrated to be associated with most human solid tumors. Both transcriptional and post-transcriptional mechanisms have been shown to modulate VEGF expression in a multitude of cell types. Here we report that when protein kinase C (PKC) pathways were activated in human glioblastoma U373 cells by phorbol 12-myristate 13-acetate (PMA), VEGF mRNA expression was up-regulated via a post-transcriptional mRNA stabilization mechanism. PMA treatment exhibited no increase in VEGF-specific transcriptional activation as determined by run-off transcription assays and VEGF promoter-luciferase reporter assays. However, PMA increased VEGF mRNA half-life from 0.8 to 3.6 h which was blocked by PKC inhibitors but not by protein kinase A or cyclic nucleotide-dependent protein kinase inhibitors. When U373 cells were transfected with antisense oligonucleotide sequences to the translation start sites of PKC-alpha, -beta, -gamma, -delta, -epsilon, or -zeta isoforms, both PKC-alpha and -zeta antisense oligonucleotides showed substantial inhibition of PMA-induced VEGF mRNA. In addition, overexpression of PKC-zeta resulted in a strong constitutive up-regulation of VEGF mRNA expression. This study demonstrates for the first time that specific PKC isoforms regulate VEGF mRNA expression through post-transcriptional mechanisms.
Collapse
Affiliation(s)
- S C Shih
- Departments of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | |
Collapse
|
44
|
Rodríguez-Martín E, Boyano-Adánez MC, Bodega G, Martín M, Hernández C, Quin Y, Vadillo M, Arilla-Ferreiro E. Redistribution of protein kinase C isoforms in rat pancreatic acini during lactation and weaning. FEBS Lett 1999; 445:356-60. [PMID: 10094489 DOI: 10.1016/s0014-5793(99)00133-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Freshly enzymatically isolated pancreatic acini from lactating and weaning Wistar rats were used to investigate the role of protein kinase C (PKC) isoforms during these physiologically relevant pancreatic secretory and growth processes. The combination of immunoblot and immunohistochemical analysis shows that the PKC isoforms alpha, delta, and epsilon are present in pancreatic acini from control, lactating and weaning rats. A vesicular distribution of PKC-alpha, -delta, and -epsilon was detected by immunohistochemical analysis in the pancreatic acini from all the experimental groups. PKC-delta showed the strongest PKC immunoreactivity (PKC-IR). In this vesicular distribution, PKC-IR was located at the apical region of the acinar cells. No differences were observed between control, lactating and weaning rats. However, the immunoblot analysis of pancreatic PKC isoforms during lactation and weaning showed a significant translocation of PKC-delta from the cytosol to the membrane fraction when compared with control animals. Translocation of PKC isoforms (alpha, delta and epsilon) in response to 12-O-tetradecanoyl phorbol 13-acetate (TPA) 1 microM (15 min, 37 degrees C) was comparable in pancreatic acini from control, lactating and weaning rats. In the control group, a significant translocation of all the isoforms (alpha, delta and epsilon) from the cytosol to the membrane was observed. The PKC isoform most translocated by TPA was PKC-delta. In contrast, no statistically significant increase in PKC-delta translocation was detected in pancreatic acini isolated from lactating or weaning rats. These results suggest that the PKC isoforms are already translocated to the surface of the acinar cells from lactating or weaning rats. In addition, they suggest that isoform specific spatial PKC distribution and translocation occur in association with the growth response previously described in the rat exocrine pancreas during lactation and weaning.
Collapse
Affiliation(s)
- E Rodríguez-Martín
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Alcalá, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
The highly disagreeable sensation of pain results from an extraordinarily complex and interactive series of mechanisms integrated at all levels of the neuroaxis, from the periphery, via the dorsal horn to higher cerebral structures. Pain is usually elicited by the activation of specific nociceptors ('nociceptive pain'). However, it may also result from injury to sensory fibres, or from damage to the CNS itself ('neuropathic pain'). Although acute and subchronic, nociceptive pain fulfils a warning role, chronic and/or severe nociceptive and neuropathic pain is maladaptive. Recent years have seen a progressive unravelling of the neuroanatomical circuits and cellular mechanisms underlying the induction of pain. In addition to familiar inflammatory mediators, such as prostaglandins and bradykinin, potentially-important, pronociceptive roles have been proposed for a variety of 'exotic' species, including protons, ATP, cytokines, neurotrophins (growth factors) and nitric oxide. Further, both in the periphery and in the CNS, non-neuronal glial and immunecompetent cells have been shown to play a modulatory role in the response to inflammation and injury, and in processes modifying nociception. In the dorsal horn of the spinal cord, wherein the primary processing of nociceptive information occurs, N-methyl-D-aspartate receptors are activated by glutamate released from nocisponsive afferent fibres. Their activation plays a key role in the induction of neuronal sensitization, a process underlying prolonged painful states. In addition, upon peripheral nerve injury, a reduction of inhibitory interneurone tone in the dorsal horn exacerbates sensitized states and further enhance nociception. As concerns the transfer of nociceptive information to the brain, several pathways other than the classical spinothalamic tract are of importance: for example, the postsynaptic dorsal column pathway. In discussing the roles of supraspinal structures in pain sensation, differences between its 'discriminative-sensory' and 'affective-cognitive' dimensions should be emphasized. The purpose of the present article is to provide a global account of mechanisms involved in the induction of pain. Particular attention is focused on cellular aspects and on the consequences of peripheral nerve injury. In the first part of the review, neuronal pathways for the transmission of nociceptive information from peripheral nerve terminals to the dorsal horn, and therefrom to higher centres, are outlined. This neuronal framework is then exploited for a consideration of peripheral, spinal and supraspinal mechanisms involved in the induction of pain by stimulation of peripheral nociceptors, by peripheral nerve injury and by damage to the CNS itself. Finally, a hypothesis is forwarded that neurotrophins may play an important role in central, adaptive mechanisms modulating nociception. An improved understanding of the origins of pain should facilitate the development of novel strategies for its more effective treatment.
Collapse
Affiliation(s)
- M J Millan
- Institut de Recherches Servier, Psychopharmacology Department, Paris, France
| |
Collapse
|
46
|
Dwivedi Y, Pandey GN. Administration of dexamethasone up-regulates protein kinase C activity and the expression of gamma and epsilon protein kinase C isozymes in the rat brain. J Neurochem 1999; 72:380-7. [PMID: 9886091 DOI: 10.1046/j.1471-4159.1999.0720380.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Altered hypothalamic-pituitary-adrenal (HPA) function (increased plasma cortisol level) has been shown to be associated with mood and behavior. Protein kinase C (PKC), an important component of the phosphatidyl-inositol signal transduction system, plays a major role in mediating various physiological functions. The present study investigates the effects of acute (single) and repeated (10-day) administrations of 0.5 or 1.0 mg/kg doses of dexamethasone (DEX), a synthetic glucocorticoid, on Bmax and KD of [3H]phorbol 12,13-dibutyrate ([3H]PDBu) binding, PKC activity, and protein expression of PKC isozymes alpha, beta, gamma, delta, and epsilon in the membrane and the cytosolic fractions of rat cortex and hippocampus. It was observed that repeated administration of 1.0 mg/kg DEX for 10 days caused a significant increase in Bmax of [3H]PDBu binding to PKC, in PKC activity, and in expressed protein levels of the gamma and epsilon isozymes in both the cytosolic and the membrane fractions of the cortex and the hippocampus, whereas a lower dose of DEX (0.5 mg/kg for 10 days) caused these changes only in the hippocampus. On the other hand, a single administration of DEX (0.5 or 1.0 mg/kg) had no significant effect on PKC in the cortex or in the hippocampus. These results suggest that alterations in HPA function from repeated administration of glucocorticoids may modulate PKC-mediated functions.
Collapse
Affiliation(s)
- Y Dwivedi
- The Psychiatric Institute, Department of Psychiatry, University of Illinois at Chicago, 60612, USA
| | | |
Collapse
|
47
|
D'Santos CS, Clarke JH, Divecha N. Phospholipid signalling in the nucleus. Een DAG uit het leven van de inositide signalering in de nucleus. BIOCHIMICA ET BIOPHYSICA ACTA 1998; 1436:201-32. [PMID: 9838115 DOI: 10.1016/s0005-2760(98)00146-5] [Citation(s) in RCA: 113] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Diverse methodologies, ranging from activity measurements in various nuclear subfractions to electron microscopy, have been used to demonstrate and establish that many of the key lipids and enzymes responsible for the metabolism of inositol lipids are resident in nuclei. PtdIns(4)P, PtdIns(4,5)P2 and PtdOH are all present in nuclei, as well as the corresponding enzyme activities required to synthesise and metabolise these compounds. In addition other non-inositol containing phospholipids such as phosphatidylcholine constitute a significant percentage of the total nuclear phospholipid content. We feel that it is pertinent to include this lipid in our discussion as it provides an alternative source of 1, 2-diacylglycerol (DAG) in addition to the hydrolysis of PtdIns(4, 5)P2. We discuss at length data related to the sources and possible consequences of nuclear DAG production as this lipid appears to be increasingly central to a number of general physiological functions. Data relating to the existence of alternative pathways of inositol phospholipid synthesis, the role of 3-phosphorylated inositol lipids and lipid compartmentalisation and transport are reviewed. The field has also expanded to a point where we can now also begin to address what role these lipids play in cellular proliferation and differentiation and hopefully provide avenues for further research.
Collapse
Affiliation(s)
- C S D'Santos
- The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | |
Collapse
|
48
|
Boris-Möller F, Wieloch T. The effect of 4 beta-phorbol-12,13-dibutyrate and staurosporine on the extracellular glutamate levels during ischemia in the rat striatum. MOLECULAR AND CHEMICAL NEUROPATHOLOGY 1998; 35:133-47. [PMID: 10343975 DOI: 10.1007/bf02815120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Hypothermia diminishes the ischemia-induced protein kinase C (PKC) translocation and inhibition, and also reduces transmitter release during ischemia. To study the role of PKC in the mechanism of glutamate release during ischemia, we measured extracellular glutamate levels in the striatum with the microdialysis technique, in the presence and absence in the dialysate of the PKC activator 4 beta-phorbol-12,13-dibutyrate (PDBu) and the protein kinase inhibitor staurosporine. We confirm that hypothermia attenuates the elevation of extracellular levels of glutamate in the striatum during ischemia. In the presence of PDBu, the glutamate levels in the dialysate increased from 0.3 mumol/L to an end ischemic level of 4.8 mumol/L during hypothermic ischemia (33 degrees C). These levels were significantly higher than in hypothermic ischemia (33 degrees C) without added PDBu. Staurosporine significantly mitigated the glutamate levels during normothermic ischemia. Our data suggest that PKC is involved in the temperature-dependent elevations of extracellular glutamate levels in the striatum during ischemia, and we propose that compounds preventing PKC activation may mimic the hypothermic protective action against ischemic brain damage.
Collapse
Affiliation(s)
- F Boris-Möller
- Laboratory for Experimental Brain Research, Wallenberg Neuroscience Center, Lund, Sweden.
| | | |
Collapse
|
49
|
Shimohama S, Saitoh T. Differential expression of protein kinase C -alpha and -betaII in rat septum and changes following fimbria-fornix lesion. Brain Res 1998; 781:343-7. [PMID: 9507185 DOI: 10.1016/s0006-8993(97)01362-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Protein kinase C (PKC)-alpha and -betaII are expressed specifically and differentially in the septal formation. Following unilateral fimbria-fornix lesions, there was a marked reduction in punctiform PKC-alpha immunoreactivity in the lateral septum and in the number of PKC-betaII immunoreactive neurons in the medial septum and the lateral septum, while some PKC-alpha and PKC-betaII immunoreactive glia-like cells were observed in the lateral septum. The response of each PKC isozyme to this lesion supports the suggested role for PKC in the plasticity of the central nervous system.
Collapse
Affiliation(s)
- S Shimohama
- Department of Neurology, Faculty of Medicine, Kyoto University, Sakyoku, Kyoto 606, Japan.
| | | |
Collapse
|