1
|
Gomez HM, Haw TJ, Ilic D, Robinson P, Donovan C, Croft AJ, Vanka KS, Small E, Carroll OR, Kim RY, Mayall JR, Beyene T, Palanisami T, Ngo DTM, Zosky GR, Holliday EG, Jensen ME, McDonald VM, Murphy VE, Gibson PG, Horvat JC. Landscape fire smoke airway exposure impairs respiratory and cardiac function and worsens experimental asthma. J Allergy Clin Immunol 2024; 154:209-221.e6. [PMID: 38513838 DOI: 10.1016/j.jaci.2024.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 02/13/2024] [Accepted: 02/22/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Millions of people are exposed to landscape fire smoke (LFS) globally, and inhalation of LFS particulate matter (PM) is associated with poor respiratory and cardiovascular outcomes. However, how LFS affects respiratory and cardiovascular function is less well understood. OBJECTIVE We aimed to characterize the pathophysiologic effects of representative LFS airway exposure on respiratory and cardiac function and on asthma outcomes. METHODS LFS was generated using a customized combustion chamber. In 8-week-old female BALB/c mice, low (25 μg/m3, 24-hour equivalent) or moderate (100 μg/m3, 24-hour equivalent) concentrations of LFS PM (10 μm and below [PM10]) were administered daily for 3 (short-term) and 14 (long-term) days in the presence and absence of experimental asthma. Lung inflammation, gene expression, structural changes, and lung function were assessed. In 8-week-old male C57BL/6 mice, low concentrations of LFS PM10 were administered for 3 days. Cardiac function and gene expression were assessed. RESULTS Short- and long-term LFS PM10 airway exposure increased airway hyperresponsiveness and induced steroid insensitivity in experimental asthma, independent of significant changes in airway inflammation. Long-term LFS PM10 airway exposure also decreased gas diffusion. Short-term LFS PM10 airway exposure decreased cardiac function and expression of gene changes relating to oxidative stress and cardiovascular pathologies. CONCLUSIONS We characterized significant detrimental effects of physiologically relevant concentrations and durations of LFS PM10 airway exposure on lung and heart function. Our study provides a platform for assessment of mechanisms that underpin LFS PM10 airway exposure on respiratory and cardiovascular disease outcomes.
Collapse
Affiliation(s)
- Henry M Gomez
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Tatt J Haw
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, Newcastle, Australia; College of Health, Medicine, and Wellbeing, Centre of Excellence Newcastle Cardio-Oncology Research Group, University of Newcastle, Callaghan, Newcastle, Australia
| | - Dusan Ilic
- Newcastle Institute for Energy and Resources, University of Newcastle, Callaghan, Australia
| | - Peter Robinson
- Newcastle Institute for Energy and Resources, University of Newcastle, Callaghan, Australia
| | - Chantal Donovan
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia; School of Life Sciences, University of Technology Sydney, Faculty of Science, Sydney, Australia
| | - Amanda J Croft
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, Newcastle, Australia; College of Health, Medicine, and Wellbeing, Centre of Excellence Newcastle Cardio-Oncology Research Group, University of Newcastle, Callaghan, Newcastle, Australia
| | - Kanth S Vanka
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia; Newcastle Institute for Energy and Resources, University of Newcastle, Callaghan, Australia
| | - Ellen Small
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Olivia R Carroll
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Richard Y Kim
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia; School of Life Sciences, University of Technology Sydney, Faculty of Science, Sydney, Australia
| | - Jemma R Mayall
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Tesfalidet Beyene
- School of Medicine and Public Health, University of Newcastle and Asthma and Breathing Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Thava Palanisami
- Global Innovative Centre for Advanced Nanomaterials, University of Newcastle, Callaghan, Australia
| | - Doan T M Ngo
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, Newcastle, Australia; College of Health, Medicine, and Wellbeing, Centre of Excellence Newcastle Cardio-Oncology Research Group, University of Newcastle, Callaghan, Newcastle, Australia
| | - Graeme R Zosky
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, Australia; College of Health and Medicine, Tasmanian School of Medicine, University of Tasmania, Hobart, Australia
| | - Elizabeth G Holliday
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
| | - Megan E Jensen
- School of Medicine and Public Health, University of Newcastle and Asthma and Breathing Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Vanessa M McDonald
- School of Medicine and Public Health, University of Newcastle and Asthma and Breathing Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Vanessa E Murphy
- School of Medicine and Public Health, University of Newcastle and Asthma and Breathing Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Peter G Gibson
- School of Medicine and Public Health, University of Newcastle and Asthma and Breathing Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Jay C Horvat
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia.
| |
Collapse
|
2
|
Donev R, Alawam K. Alterations in Gene Expression in Depression: Prospects for Personalize Patient Treatment. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2015; 101:97-124. [PMID: 26572977 DOI: 10.1016/bs.apcsb.2015.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The number of people around the world suffering from depression has dramatically increased in last few decades. It has been predicted that by 2020 depression will become the second most common cause of disability. Furthermore, depression is often misdiagnosed and confused with other psychiatric disorders showing similar symptoms, i.e., anxiety and bipolar disorder, due to the fact that diagnosing is often carried out by medical workers who are not psychiatrically trained. These facts prompt us to prepare this review which focuses on alterations in gene expression in depression. We believe that an in-depth knowledge of molecular bases of behavior in depression and other mood disorders would be of a great benefit for the correct diagnosing of these disorders, as well as for prescribing a treatment that best suits each individual depending on expression alterations in depression-related genes. Therefore, the main aim of this review is to promote further translational research on the biochemistry of mood disorders and take the results further for the design of new targeted therapeutics that can be used for personalized treatment with minimal adverse effects.
Collapse
Affiliation(s)
| | - Khaled Alawam
- Forensic Medicine Department, Ministry of Interior, Kuwait City, Kuwait
| |
Collapse
|
3
|
Yu HR, Kuo HC, Chen CC, Sheen JM, Tiao MM, Chen YC, Chang KA, Tain YL, Huang LT. Prenatal dexamethasone exposure in rats results in long-term epigenetic histone modifications and tumour necrosis factor-α production decrease. Immunology 2015; 143:651-60. [PMID: 24962734 DOI: 10.1111/imm.12346] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 06/19/2014] [Accepted: 06/19/2014] [Indexed: 12/14/2022] Open
Abstract
Glucocorticoid (GC) is often given when preterm delivery is expected. This treatment is successful in stimulating the development of the fetal lung. However, reports and related research regarding the prolonged effects of prenatal GC on the development of immunity are very limited. Some data, derived from infants whose mothers were given immunosuppressants during pregnancy for the treatment of autoimmune disorders, suggest that prenatal exposure to GC may have only a limited effect on the development of the immune system. What is unknown is whether the immune modulation effects of prenatal GC might appear at a later childhood stage and beyond. Here we evaluated the immune programming influenced by prenatal GC. Pregnant Sprague-Dawley rats received dexamethasone (DEX; 0.1 mg/kg/day) or saline at gestational days 14-20. Male offspring were killed at day 7 or day 120 after birth. Spleens were collected for immune study. Of the inflammation mediators, matrix metalloproteinase-9, tumour necrosis factor-α (TNF-α) and granulocyte-macrophage colony-stimulating factor mRNAs decreased in the prenatal DEX group at an early stage after birth. Upon concanavalin A stimulation, prenatal DEX treatment reduced TNF-α production, but not interferon-γ production, by splenocytes at day 120 after birth compared with the vehicle group. Decreased levels of active chromatin signs (acetylation of histone H3 lysines, H3K4me1/3, and H3K36me3) in TNF-α promoter were compatible with the expressions of TNF-α. Our results suggest that prenatal DEX has a profound and lasting impact on the developing immune system even to the adult stage. Epigenetic histone modifications regulate TNF-α expression following prenatal DEX in rats.
Collapse
Affiliation(s)
- Hong-Ren Yu
- Department of Paediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Centre, Graduate Insititute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Aziz MH, Shen H, Maki CG. Glucocorticoid receptor activation inhibits p53-induced apoptosis of MCF10Amyc cells via induction of protein kinase Cε. J Biol Chem 2012; 287:29825-36. [PMID: 22773829 DOI: 10.1074/jbc.m112.393256] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Glucocorticoid receptor (GR) is a ligand-dependent transcription factor that can promote apoptosis or survival in a cell-specific manner. Activated GR has been reported to inhibit apoptosis in mammary epithelial cells and breast cancer cells by increasing pro-survival gene expression. In this study, activated GR inhibited p53-dependent apoptosis in MCF10A cells and human mammary epithelial cells that overexpress the MYC oncogene. Specifically, GR agonists hydrocortisone or dexamethasone inhibited p53-dependent apoptosis induced by cisplatin, ionizing radiation, or the MDM2 antagonist Nutlin-3. In contrast, the GR antagonist RU486 sensitized the cells to apoptosis by these agents. Apoptosis inhibition was associated with maintenance of mitochondrial membrane potential, diminished caspase-3 and -7 activation, and increased expression at both the mRNA and protein level of the anti-apoptotic PKC family member PKCε. Knockdown of PKCε via siRNA targeting reversed the protective effect of dexamethasone and restored apoptosis sensitivity. These data provide evidence that activated GR can inhibit p53-dependent apoptosis through induction of the anti-apoptotic factor PKCε.
Collapse
|
5
|
Kang M, Ryu J, Kim JH, Na H, Zuo Z, Do SH. Corticosterone decreases the activity of rat glutamate transporter type 3 expressed in Xenopus oocytes. Steroids 2010; 75:1113-8. [PMID: 20654639 DOI: 10.1016/j.steroids.2010.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Revised: 06/21/2010] [Accepted: 07/14/2010] [Indexed: 11/25/2022]
Abstract
Glucocorticoids can increase the extracellular concentrations of glutamate, the major excitatory neurotransmitter. We investigated the effects of corticosterone on the activity of a glutamate transporter, excitatory amino acid carrier 1 (EAAC1; also called excitatory amino acid transporter type 3 [EAAT3]), and the roles of protein kinase C (PKC) and phosphatidylinositol 3-kinase (PI3K) in regulating these effects. Rat EAAC1 was expressed in Xenopus oocytes by injecting mRNA. L-Glutamate (30 μM)-induced membrane currents were measured using the two-electrode voltage clamp technique. Exposure of these oocytes to corticosterone (0.01-1 μM) for 72 h decreased EAAC1 activity in a dose-dependent fashion, and this inhibition was incubation time-dependent. Corticosterone (0.01 μM for 72 h) significantly decreased the V(max), but not the K(m), of EAAC1 for glutamate. Furthermore, pretreatment of oocytes with staurosporine, a PKC inhibitor, significantly decreased EAAC1 activity (1.00±0.06 to 0.70±0.05 μC; P<0.05). However, no statistical differences were observed between oocytes treated with staurosporine, corticosterone, or corticosterone plus staurosporine. Similar patterns of responses were achieved by chelerythrine or calphostin C, other PKC inhibitors. Phorbol-12-myristate-13-acetate (PMA), a PKC activator, inhibited corticosterone-induced reduction in EAAC1 activity. Pretreating oocytes with wortmannin or LY294002, PI3K inhibitors, also significantly reduced EAAC1 activity, but no difference was observed between oocytes treated with wortmannin, corticosterone, or wortmannin plus corticosterone. The above results suggest that corticosterone exposure reduces EAAC1 activity and this effect is PKC- and PI3K-dependent.
Collapse
Affiliation(s)
- Maehwa Kang
- Department of Anesthesiology, Seoul National University College of Medicine, Seoul, South Korea
| | | | | | | | | | | |
Collapse
|
6
|
Palumbo ML, Zorrilla Zubilete MA, Cremaschi GA, Genaro AM. Different effect of chronic stress on learning and memory in BALB/c and C57BL/6 inbred mice: Involvement of hippocampal NO production and PKC activity. Stress 2009; 12:350-61. [PMID: 19006005 DOI: 10.1080/10253890802506383] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Nitric oxide (NO) has been involved in many pathophysiological brain processes. Recently, we showed that neuronal nitric oxide synthase (nNOS)-mediated decrease in NO production is involved in memory impairment induced by chronic mild stress (CMS) in BALB/c mice. Two genetically different inbred murine strains, C57BL/6 and BALB/c, show distinct behavioral responses, neurodevelopmental and neurochemical parameters. Here, we perform a comparative study on CMS effects upon learning and memory in both strains, analyzing the role of NO production and its regulation by protein kinase C (PKC). Stressed BALB/c, but not C57Bl/6 mice, showed a poor learning performance in both the open field and passive avoidance inhibitory tasks. Also, CMS induced a diminished NO production by nNOS, associated with an increment in gamma and zeta PKC isoenzymes in BALB/c mice. In C57BL/6 mice, CMS had no effect on NO production, but increased delta and decreased betaI PKC isoforms. In vivo administration of a NOS inhibitor induced behavioral alterations in both strains. These results suggest a differential effect of stress, with BALB/c being more vulnerable to stress than C57BL/6 mice. This effect could be related to a differential regulation of NOS and PKC isoenzymes, pointing to an important role of NO in learning and memory.
Collapse
Affiliation(s)
- María Laura Palumbo
- CEFYBO-CONICET, 1a Cát de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | | | | |
Collapse
|
7
|
Zarate J, Churruca I, Echevarría E, Casis L, López de Jesús M, Saenz del Burgo L, Sallés J. Immunohistochemical localization of CB1 cannabinoid receptors in frontal cortex and related limbic areas in obese Zucker rats: effects of chronic fluoxetine treatment. Brain Res 2008; 1236:57-72. [PMID: 18722357 DOI: 10.1016/j.brainres.2008.07.100] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2008] [Revised: 07/17/2008] [Accepted: 07/18/2008] [Indexed: 11/17/2022]
Abstract
In the present study, we report on the application of two specific polyclonal antibodies to different intracellular domains of the CB1 cannabinoid receptor to define the expression of the neural CB1 cannabinoid receptor at the histochemical level in frontal cortex and related limbic areas of the obese Zucker rats. Higher levels of CB1 receptor expression in frontal, cingulated and piriform cortex, without differences in temporal, parietal and occipital cortex, were observed in obese Zucker rats, with respect to their lean littermates. CB1 phosphorylated receptor (CB1-P) levels were also higher in frontal, temporal, parietal and occipital cortex in obese rats with respect to lean controls. Potential involvement of brain cortical CB1 cannabinoid receptors in the long-term effects of fluoxetine was studied. Experimental animals were administered with fluoxetine (10 mg/kg, i.p.) daily for 3 weeks, whereas the control group was given 0.9% NaCl solution. In obese Zucker rats, a significant decrease in CB1 receptor levels, measured by western blot, was observed in brain cortex after fluoxetine treatment. Immunostaining for CB1 receptor expression was also carried out, showing a significant decrease in the density of neural cells positive for CB1 receptor in frontal, cingulate and piriform cortex, without changes in parietal, temporal and occipital regions. Regional prosencephalic immunostaining for CB1-P receptor level showed a significant decrease in the density of stained neural cells in frontal, temporal and parietal cortex, without changes in cingulated, piriform and occipital cortex. These results suggest the involvement of endocannabinoid system in the chronic effects of fluoxetine, especially in the frontal cortex.
Collapse
Affiliation(s)
- J Zarate
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country, Paseo de la Universidad 7, 01006 Vitoria, Spain
| | | | | | | | | | | | | |
Collapse
|
8
|
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
|
9
|
Palumbo ML, Fosser NS, Rios H, Zorrilla Zubilete MA, Guelman LR, Cremaschi GA, Genaro AM. Loss of hippocampal neuronal nitric oxide synthase contributes to the stress-related deficit in learning and memory. J Neurochem 2007; 102:261-74. [PMID: 17419805 DOI: 10.1111/j.1471-4159.2007.04528.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Nitric oxide (NO) has been involved in many pathophysiological brain processes. However, the exact role of NO in the cognitive deficit associated to chronic stress exposure has not been elucidated. In this study, we investigated the participation of hippocampal NO production and their regulation by protein kinase C (PKC) in the memory impairment induced in mice subjected to chronic mild stress model (CMS). CMS mice showed a poor learning performance in both open field and passive avoidance inhibitory task respect to control mice. Histological studies showed a morphological alteration in the hippocampus of CMS mice. On the other hand, chronic stress induced a diminished NO production by neuronal nitric oxide synthase (nNOS) correlated with an increment in gamma and zeta PKC isoenzymes. Partial restoration of nNOS activity was obtained after PKC activity blockade. NO production by inducible nitric oxide synthase isoform was not detected. The magnitude of oxidative stress, evaluated by reactive oxygen species production, after excitotoxic levels of NMDA was increased in hippocampus of CMS mice. Moreover, ROS formation was higher in the presence of nNOS inhibitor in both control and CMS mice. Finally, treatment of mice with nNOS inhibitors results in behavioural alterations similar to those observed in CMS animals. These findings suggest a novel role for nNOS showing protective activity against insults that trigger tissue toxicity leading to memory impairments.
Collapse
Affiliation(s)
- María Laura Palumbo
- CEFYBO-CONICET and 1a. Cát. de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
10
|
Zhu XY, Liu YJ, Diao F, Fan J, Lu J, Xu RB. Role of glucocorticoids and glucocorticoid receptor in priming of macrophages caused by glucocorticoid receptor blockade. Endocrine 2007; 31:130-7. [PMID: 17873323 DOI: 10.1007/s12020-007-0019-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2007] [Revised: 05/10/2007] [Accepted: 05/14/2007] [Indexed: 11/29/2022]
Abstract
We previously reported that glucocorticoid receptor (GR) blockade (injected with GR antagonist RU486) primed the host responses to lipopolysaccharide. Since decrease of GR and elevated glucocorticoids (GCs) have been always reported as parallel responses, we hypothesize that both GCs and GR play important roles in GR blockade induced priming. We first confirm that the production of nitric oxide (NO), superoxide (O2-), and PKCalpha expression are all increased in peritoneal macrophages from GR blockade rats, indicating that macrophages are primed by GR blockade. Furthermore, using unilateral adrenalectomy rats, we find that the elevated GCs caused by a feedback mechanism following GR blockade may be involved in the process of priming. In vitro experiments in RAW264.7 cells show the inhibitory effect of GCs on NO production, which can be thoroughly blocked by RU486, indicating the increase of NO production in GR blockade rats is due to the elimination of GCs's anti-inflammatory function. In contrast, 10(-7) M corticosterone induces significant increases in O2- release, PKCalpha expression and phosphorylation, which cannot be reversed by RU486, demonstrating a previously unrecognized pro-inflammatory role of GCs in enhancing PM activation through a GR-independent pathway. The effect of GCs on PKCalpha expression even exists in GR deficient COS-7 cells as well as in GR knock-down RAW264.7 cells. In conclusion, both GR impairment and elevation of GCs are involved in the priming of macrophages caused by GR blockade. The findings of the divergent roles of GCs in modulation of inflammation may change therapeutic strategy for inflammatory diseases with GCs.
Collapse
Affiliation(s)
- Xiao-Yan Zhu
- Department of Physiology, Second Millitary Medical University, 800 Xiang Yin Road, Shanghai, 200433, PR China.
| | | | | | | | | | | |
Collapse
|
11
|
Rausch JL. Initial conditions of psychotropic drug response: studies of serotonin transporter long promoter region (5-HTTLPR), serotonin transporter efficiency, cytokine and kinase gene expression relevant to depression and antidepressant outcome. Prog Neuropsychopharmacol Biol Psychiatry 2005; 29:1046-61. [PMID: 16005136 DOI: 10.1016/j.pnpbp.2005.03.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/01/2005] [Indexed: 12/29/2022]
Abstract
The Hypothesis of Initial Conditions posits that differences in psychotropic drug response result from individual differences in receptor site kinetics, and differences in the sensitivity of downstream receptor-linked responses. This work examines data consistent with the hypothesis, specific to genetic and kinetic differences of the serotonin (5-HT) transporter (SERT), as they may be linked to divergent antidepressant response (ADR). The mechanisms for divergent ADR in association with different initial SERT function are considered within the context of SERT trafficking as sensitive to various different kinase and cytokine signals, some of which are dependent on the 5-HTTLPR polymorphism of the SERT gene. Pilot data suggest that human lymphocytes show kinase changes similar to those found in rat brain with ADT. These studies additionally suggest that ADT prompts a shift in cytokine gene expression toward a greater anti-inflammatory/inflammatory ratio. These latter findings are discussed within the context of a literature suggesting increased inflammatory cytokine levels in depression, and recent observations of increased temperature associated with depression. In sum, the data suggest the opportunity to identify response dependent protein (RDP) expression patterns that may differ with dichotomous ADR, and suggest new insights into understanding the mechanisms of psychotropic drug response through an understanding of initial differences in potential for psychotropic drug target regulation during therapy.
Collapse
Affiliation(s)
- Jeffrey L Rausch
- Veterans Administration, Department of Psychiatry and Health Behavior, The Medical College of Georgia, Augusta, GA 30912, USA.
| |
Collapse
|
12
|
Bíró T, Griger Z, Kiss E, Papp H, Aleksza M, Kovács I, Zeher M, Bodolay E, Csépány T, Szûcs K, Gergely P, Kovács L, Szegedi G, Sipka S. Abnormal Cell-Specific Expressions of Certain Protein Kinase C Isoenzymes in Peripheral Mononuclear Cells of Patients with Systemic Lupus Erythematosus: Effect of Corticosteroid Application. Scand J Immunol 2004; 60:421-8. [PMID: 15379867 DOI: 10.1111/j.0300-9475.2004.01485.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We have studied the expressions of various protein kinase C (PKC) isoenzymes in T cells and monocytes from patients with systemic lupus erythematosus (SLE), in comparison to those of healthy controls and patients with other immunological disorders. As measured by Western blotting, the levels of PKCbeta, delta, eta, epsilon, theta and zeta (but not of PKCalpha) significantly decreased in T cells of SLE patients. In monocytes, however, we observed marked suppressions only in the expressions of PKCdelta, epsilon and zeta but not in the expressions of other PKC isoforms. In vivo corticosteroid application, as well as in vitro steroid treatment of monocytes, elevated the expressions of most isoforms close to normal values; however, the decreased levels of PKCtheta and zeta were not affected by steroid application. These alterations were characteristic to SLE because we could not detect any changes in the PKC levels in mononuclear cells of primary Sjögren's syndrome and mixed connective tissue disease patients. These results suggest that impaired PKC isoenzyme pattern may exist in the T cells and monocytes of SLE patients. Furthermore, the clinically efficient glucocorticoid application in SLE can increase the expression of some members of PKC system.
Collapse
Affiliation(s)
- T Bíró
- Department of Physiology and Cell Physiology Research Group of the Hungarian Academy of Sciences, Debrecen, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
McNamara RK, Lenox RH. Acute restraint stress reduces protein kinase Cγ in the hippocampus of C57BL/6 but not DBA/2 mice. Neurosci Lett 2004; 368:293-6. [PMID: 15364414 DOI: 10.1016/j.neulet.2004.07.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2004] [Revised: 06/11/2004] [Accepted: 07/14/2004] [Indexed: 11/26/2022]
Abstract
Protein kinase C gamma (PKC gamma) is highly expressed in the rodent hippocampus and has been implicated in long-term alterations in synaptic efficacy. Acute stress has been shown to negatively affect hippocampal synaptic plasticity, and the present study examined the effect of acute stress on PKC gamma expression/subcellular distribution by quantitative western blotting in two inbred mouse strains (C57BL/6J versus DBA/2J) with established differences in hippocampal plasticity. It was found that both DBA/2J and C57BL/6J strains exhibited similar basal, stress-induced elevations, and recovery of serum corticosterone levels. Acute stress produced a significant reduction in both membrane and cytosolic PKC gamma expression in the hippocampus of C57BL/6J mice compared to no-stress controls, but did not alter either membrane or cytosolic PKC gamma expression in the hippocampus of DBA/2J mice compared to no-stress controls. These data provide direct evidence that PKC gamma is differentially regulated in the hippocampus of C57BL/6J and DBA/2J mice by acute stress. The role of stress-induced regulation of hippocampal PKC gamma expression in hippocampal synaptic plasticity is discussed.
Collapse
Affiliation(s)
- Robert K McNamara
- Department of Psychiatry, University of Cincinnati College of Medicine, Medical Science Building, 231 Albert Sabin Way, Cincinnati, OH 45267-0559, USA.
| | | |
Collapse
|
14
|
Gould TD, Quiroz JA, Singh J, Zarate CA, Manji HK. Emerging experimental therapeutics for bipolar disorder: insights from the molecular and cellular actions of current mood stabilizers. Mol Psychiatry 2004; 9:734-55. [PMID: 15136794 DOI: 10.1038/sj.mp.4001518] [Citation(s) in RCA: 165] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bipolar disorder afflicts approximately 1-3% of both men and women, and is coincident with major economic, societal, medical, and interpersonal consequences. Current mediations used for its treatment are associated with variable rates of efficacy and often intolerable side effects. While preclinical and clinical knowledge in the neurosciences has expanded at a tremendous rate, recent years have seen no major breakthroughs in the development of novel types of treatment for bipolar disorder. We review here approaches to develop novel treatments specifically for bipolar disorder. Deliberate (ie not by serendipity) treatments may come from one of two general mechanisms: (1) Understanding the mechanism of action of current medications and thereafter designing novel drugs that mimics these mechanism(s); (2) Basing medication development upon the hypothetical or proven underlying pathophysiology of bipolar disorder. In this review, we focus upon the first approach. Molecular and cellular targets of current mood stabilizers include lithium inhibitable enzymes where lithium competes for a magnesium binding site (inositol monophosphatase, inositol polyphosphate 1-phosphatase, glycogen synthase kinase-3 (GSK-3), fructose 1,6-bisphosphatase, bisphosphate nucleotidase, phosphoglucomutase), valproate inhibitable enzymes (succinate semialdehyde dehydrogenase, succinate semialdehyde reductase, histone deacetylase), targets of carbamazepine (sodium channels, adenosine receptors, adenylate cyclase), and signaling pathways regulated by multiple drugs of different classes (phosphoinositol/protein kinase C, cyclic AMP, arachidonic acid, neurotrophic pathways). While the task of developing novel medications for bipolar disorder is truly daunting, we are hopeful that understanding the mechanism of action of current mood stabilizers will ultimately lead clinical trials with more specific medications and thus better treatments those who suffer from this devastating illness.
Collapse
Affiliation(s)
- T D Gould
- Laboratory of Molecular Pathophysiology, National Institute of Mental Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
15
|
McNamara RK, Vasquez PA, Mathe AA, Lenox RH. Differential expression and regulation of myristoylated alanine-rich C kinase substrate (MARCKS) in the hippocampus of C57/BL6J and DBA/2J mice. J Neurochem 2003; 85:462-8. [PMID: 12675922 DOI: 10.1046/j.1471-4159.2003.01700.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The myristoylated alanine-rich C kinase substrate (MARCKS) is a major protein kinase C (PKC) substrate in brain that binds the inner surface of the plasma membrane, calmodulin, and cross-links filamentous actin, all in a PKC phosphorylation-reversible manner. MARCKS has been implicated in hippocampal-dependent learning and long-term potentiation (LTP). Previous studies have shown DBA/2 mice to exhibit poor spatial/contextual learning, impaired hippocampal LTP, and hippocampal mossy fiber hypoplasia, as well as reduced hippocampal PKC activity and expression relative to C57BL/6 mice. In the present study, we assessed the expression (mRNA and protein) and subcellular distribution (membrane and cytolsol) of MARCKS in the hippocampus and frontal cortex of C57BL/6 and DBA/2 mice using quantitative western blotting. In the hippocampus, total MARCKS mRNA and protein levels in C57BL/6J mice were significantly lower ( approximately 45%) compared with DBA/2J mice, and MARCKS protein was observed predominantly in the cytosolic fraction. MARCKS expression in frontal cortex did not differ significantly between strains. To examine the dynamic regulation of MARCKS subcellular distribution, mice from each strain were subjected to 60 min restraint stress and MARCKS subcellular distribution was determined 24 h later. Restraint stress resulted in a significant reduction in membrane MARCKS expression in C57BL/6J hippocampus but not in the DBA/2J hippocampus despite similar stress-induced increases in serum corticosterone. Restraint stress did not affect cytosolic or total MARCKS levels in either strain. Similarly, restraint stress (30 min) in rats also induced a significant reduction in membrane MARCKS, but not total or cytosolic MARCKS, in the hippocampus but not in frontal cortex. In rats, chronic lithium treatment prior to stress exposure reduced hippocampal MARCKS expression but did not affect the stress-induced reduction in membrane MARCKS. Collectively these data demonstrate higher resting levels of MARCKS in the hippocampus of DBA/2J mice compared to C57BL/6J mice, and that acute stress leads to a long-term reduction in membrane MARCKS expression in C57BL/6J mice and rats but not in DBA/2J mice. These strain differences in hippocampal MARCKS expression and subcellular translocation following stress may contribute to the differences in behaviors requiring hippocampal plasticity observed between these strains.
Collapse
Affiliation(s)
- Robert K McNamara
- Department of Psychiatry, University of Pennsylvania School of Medicine, Clinical Research Building, Philadelphia, Pennsylvania, USA.
| | | | | | | |
Collapse
|
16
|
Johnston AR, Seckl JR, Dutia MB. Role of the flocculus in mediating vestibular nucleus neuron plasticity during vestibular compensation in the rat. J Physiol 2002; 545:903-11. [PMID: 12482895 PMCID: PMC2290729 DOI: 10.1113/jphysiol.2002.024281] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2002] [Accepted: 09/26/2002] [Indexed: 11/08/2022] Open
Abstract
We investigated the role of the cerebellar flocculus in mediating the adaptive changes that occur in the intrinsic properties of brainstem medial vestibular nucleus (MVN) neurons during vestibular compensation. Ipsi-lesional, but not contra-lesional, flocculectomy prevented the compensatory increase in intrinsic excitability (CIE) that normally occurs in the de-afferented MVN neurons within 4 h after unilateral labyrinthectomy (UL). Flocculectomy did not, however, prevent the down-regulation of efficacy of GABA receptors that also occurs in these neurons after UL, indicating that these responses of the MVN neurons to deafferentation are discrete, parallel processes. CIE was also abolished by intra-floccular microinjection of the metabotropic glutamate receptor (mGluR) antagonist AIDA, and the protein kinase C inhibitor bisindolymaleimide I (BIS-I). The serene-threonine kinase inhibitor H-7 had no effect when microinjected at the time of de-afferentation, but abolished CIE if microinjected 2 h later. These cellular effects are in line with the recently reported retardatory effects of BIS-I and H-7 on behavioural recovery after UL. They demonstrate that the increase in intrinsic excitability in MVN neurons during vestibular compensation is cerebellum dependent, and requires mGluR activation and protein phosphorylation in cerebellar cortex. Furthermore, microinjection of the glucocorticoid receptor (GR) antagonist RU38486 into the ipsi-lesional flocculus also abolished CIE in MVN neurons. Thus an important site for glucocorticoids in facilitating vestibular compensation is within the cerebellar cortex. These observations ascribe functional significance to the high levels of GR and 11-beta-HSD Type 1 expression in cerebellum.
Collapse
Affiliation(s)
- Alex R Johnston
- Department of Biomedical Sciences (Physiology), Edinburgh University Medical School, Hugh Robson Building, George Square, Edinburgh EH8 9XD, Scotland, UK
| | | | | |
Collapse
|
17
|
Abstract
Mood stabilizers represent a class of drugs that are efficacious in the treatment of bipolar disorder. The most established medications in this class are lithium, valproic acid, and carbamazepine. In addition to their therapeutic effects for treatment of acute manic episodes, these medications often are useful as prophylaxis against future episodes and as adjunctive antidepressant medications. While important extracellular effects have not been excluded, most available evidence suggests that the therapeutically relevant targets of this class of medications are in the interior of cells. Herein we give a prospective of a rapidly evolving field, discussing common effects of mood stabilizers as well as effects that are unique to individual medications. Mood stabilizers have been shown to modulate the activity of enzymes, ion channels, arachidonic acid turnover, G protein coupled receptors and intracellular pathways involved in synaptic plasticity and neuroprotection. Understanding the therapeutic targets of mood stabilizers will undoubtedly lead to a better understanding of the pathophysiology of bipolar disorder and to the development of improved therapeutics for the treatment of this disease. Furthermore, the involvement of mood stabilizers in pathways operative in neuroprotection suggests that they may have utility in the treatment of classical neurodegenerative disorders.
Collapse
Affiliation(s)
- Todd D. Gould
- Laboratory of Molecular Pathophysiology, Building 49, Room B1EE16, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Guang Chen
- Laboratory of Molecular Pathophysiology, Building 49, Room B1EE16, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Husseini K. Manji
- Laboratory of Molecular Pathophysiology, Building 49, Room B1EE16, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Perez J, Tardito D, Racagni G, Smeraldi E, Zanardi R. cAMP signaling pathway in depressed patients with psychotic features. Mol Psychiatry 2002; 7:208-12. [PMID: 11840314 DOI: 10.1038/sj.mp.4000969] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2001] [Revised: 07/13/2001] [Accepted: 07/15/2001] [Indexed: 11/08/2022]
Abstract
Abnormalities in protein kinase A (PKA) and Rap1 have recently been reported in depressed patients. The aim of the present study was to investigate the levels of these proteins in platelets from untreated unipolar and bipolar depressed patients with psychotic features. The levels PKA and Rap1 were assessed by Western blot analysis and immunostaining in 37 drug-free patients and 29 healthy subjects. Both unipolar and bipolar patients with psychotic depression have significantly lower levels of platelet regulatory type I and higher levels of catalytic subunits of PKA than controls, whereas the levels of regulatory type II were higher only in psychotic unipolar patients. No significant differences were found in the immunolabeling of both Rap1 and actin among groups. These findings support the idea that besides nonpsychotic depression, abnormalities of PKA could be linked, albeit in a somewhat different way, with psychotic depression.
Collapse
Affiliation(s)
- J Perez
- Istituto Scientifico HSR, Department of Neuropsychiatry, School of Medicine, University Vita-Salute, Milan, Italy.
| | | | | | | | | |
Collapse
|
19
|
Chen G, Masana MI, Manji HK. Lithium regulates PKC-mediated intracellular cross-talk and gene expression in the CNS in vivo. Bipolar Disord 2000; 2:217-36. [PMID: 11249800 DOI: 10.1034/j.1399-5618.2000.20303.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
It has become increasingly appreciated that the long-term treatment of complex neuropsychiatric disorders like bipolar disorder (BD) involves the strategic regulation of signaling pathways and gene expression in critical neuronal circuits. Accumulating evidence from our laboratories and others has identified the family of protein kinase C (PKC) isozymes as a shared target in the brain for the long-term action of both lithium and valproate (VPA) in the treatment of BD. In rats chronically treated with lithium at therapeutic levels, there is a reduction in the levels of frontal cortical and hippocampal membrane-associated PKC alpha and PKC epsilon. Using in vivO microdialysis, we have investigated the effects of chronic lithium on the intracellular cross-talk between PKC and the cyclic AMP (cAMP) generating system in vivo. We have found that activation of PKC produces an increase in dialysate cAMP levels in both prefrontal cortex and hippocampus, effects which are attenuated by chronic lithium administration. Lithium also regulates the activity of another major signaling pathway the c-Jun N-terminal kinase pathway--in a PKC-dependent manner. Both Li and VPA, at therapeutically relevant concentrations, increase the DNA binding of activator protein 1 (AP-1) family of transcription factors in cultured cells in vitro, and in rat brain ex vivo. Furthermore, both agents increase the expression of an AP-1 driven reporter gene, as well as the expression of several endogenous genes known to be regulated by AP-1. Together, these results suggest that the PKC signaling pathway and PKC-mediated gene expression may be important mediators of lithium's long-term therapeutic effects in a disorder as complex as BD.
Collapse
Affiliation(s)
- G Chen
- Department of Psychiatry and Behavioral Neurosciences, WSU School of Medicine, Detroit, MI 48201, USA.
| | | | | |
Collapse
|
20
|
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
|
21
|
Xan HJ, Kim DH, Park SH. Regulatory mechanisms of Na/Pi cotransporter by glucocorticoid in renal proximal tubule cells: involvement of cAMP and PKC. Kidney Blood Press Res 1999; 23:1-9. [PMID: 10567847 DOI: 10.1159/000025947] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The signaling pathways involved in the regulation of glucocorticoid on Pi uptake were examined in primary cultured rabbit renal proximal tubule cells (PTCs). Dexamethasone (DEX, 10(-9) M) inhibited Pi uptake, although aldosterone, a mineralocorticoid, did not affect Pi uptake. Its effect was due to a 23% decrease in the V(max) value. DEX-induced inhibition of Pi uptake was prevented by actinomycin D, cycloheximide, and the glucocorticoid receptor antagonists, progesterone and cortexolone. SQ 22536 (adenylate cyclase inhibitor) and the myristoylated protein kinase A inhibitor amide 14-22 (PKI) did not block the DEX-induced inhibition of Pi uptake. Indeed, DEX did not affect cAMP production. However, neomycin and U 73122 (PLC inhibitors), staurosporine and bisindolylmaleimide I (PKC inhibitors) blocked the DEX-induced inhibition of Pi uptake. In addition, DEX increased the membrane-bound PKC activity from 2. 82+/-0.21 to 4.16+/-0.34 pmol/mg protein/min. These findings demonstrate that glucocorticoid inhibits Pi uptake and its effect is genomic and receptor-mediated and the activation of the PLC/PKC pathway is involved in its effect on the PTCs.
Collapse
Affiliation(s)
- H J Xan
- Department of Veterinary Physiology, College of Veterinary Medicine, Hormone Research Center, Chonnam National University, Kwangju, Korea.
| | | | | |
Collapse
|
22
|
Manji HK, Lenox RH. Ziskind-Somerfeld Research Award. Protein kinase C signaling in the brain: molecular transduction of mood stabilization in the treatment of manic-depressive illness. Biol Psychiatry 1999; 46:1328-51. [PMID: 10578449 DOI: 10.1016/s0006-3223(99)00235-8] [Citation(s) in RCA: 168] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Understanding the biology of the pharmacological stabilization of mood will undoubtedly serve to provide significant insight into the pathophysiology of manic-depressive illness (MDI). Accumulating evidence from our laboratories and those of other researchers has identified the family of protein kinase C isozymes as a shared target in the brain for the long-term action of both lithium and valproate. In rats chronically treated with lithium, there is a reduction in the hippocampus of the expression of two protein kinase isozymes, alpha and epsilon, as well as a reduction in the expression of a major PKC substrate, MARCKS, which has been implicated in long-term neuroplastic events in the developing and adult brain. In addition, we have been investigating the down-stream impact of these mood stabilizers on another kinase system, GSK-3 beta and on the AP-1 family of transcription factors. Further studies have generated promising preliminary data in support of the antimanic action of tamoxifen, and antiestrogen that is also a PKC inhibitor. Future studies must address the therapeutic relevance of these protein targets in the brain using innovative strategies in both animal and clinical investigations to ultimately create opportunities for the discovery of the next generations of mood stabilizers for the treatment of MDI.
Collapse
Affiliation(s)
- H K Manji
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | |
Collapse
|
23
|
Dwivedi Y, Pandey GN. Repeated administration of dexamethasone increases phosphoinositide-specific phospholipase C activity and mRNA and protein expression of the phospholipase C beta 1 isozyme in rat brain. J Neurochem 1999; 73:780-90. [PMID: 10428076 DOI: 10.1046/j.1471-4159.1999.0730780.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [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 has been shown to be associated with changes in mood and behavior. The enzyme phosphoinositide-specific phospholipase C (PI-PLC), an important component of the PI signal transduction system, plays a major role in mediating various physiological functions. In the present study, we investigated the effects of a single dose and of repeated administration (0.5 or 1.0 mg/kg for 10 days) of dexamethasone (DEX), a synthetic glucocorticoid, on PI-PLC activity and on expression of PLC isozymes (beta1, delta1, and gamma1) in rat brain. Repeated administration of DEX (1.0 mg/kg) caused a significant increase in PI-PLC activity and in protein expression of the PLC beta1 isozyme in both membrane and cytosol fractions of cortex and hippocampus; however, the repeated administration of a smaller dose of DEX (0.5 mg/kg) caused these changes only in hippocampus but not in cortex. The increase in PLC beta1 protein was associated with an increase in its mRNA level, as measured by competitive RT-PCR. A single administration of DEX (0.5 or 1.0 mg/kg) to rats had no significant effects on PI-PLC activity or on the protein expression of PLC isozymes. These results suggest that DEX up-regulates PI-PLC in rat brain, which presumably is due to a selective increase in expression of the PLC beta1 isozyme, and that these changes in PI-PLC may be related to HPA axis-mediated changes in mood and behavior.
Collapse
Affiliation(s)
- Y Dwivedi
- Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, 60612, USA
| | | |
Collapse
|
24
|
Tueting P, Costa E, Dwivedi Y, Guidotti A, Impagnatiello F, Manev R, Pesold C. The phenotypic characteristics of heterozygous reeler mouse. Neuroreport 1999; 10:1329-34. [PMID: 10363948 DOI: 10.1097/00001756-199904260-00032] [Citation(s) in RCA: 158] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Histological and behavioral traits are associated with reelin (Reln) haplo-insufficiency in heterozygous reeler mouse (rl+/-). These phenotypic traits are an approximately 50% decrease of brain Reln mRNA and Reln protein, an accumulation of nicotinamide-adenine dinucleotide phosphate-diaphorase (NADPH-d)-positive neurons in subcortical white matter, an age-dependent decrease in prepulse inhibition of startle (PPI), and neophobic behavior on the elevated plus-maze. Possible analogies between these rl+/- phenotypic traits and signs of psychosis vulnerability are discussed.
Collapse
Affiliation(s)
- P Tueting
- Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, 60612, USA
| | | | | | | | | | | | | |
Collapse
|