1
|
Bernstein HG, Smalla KH, Keilhoff G, Dobrowolny H, Kreutz MR, Steiner J. The many "Neurofaces" of Prohibitins 1 and 2: Crucial for the healthy brain, dysregulated in numerous brain disorders. J Chem Neuroanat 2023; 132:102321. [PMID: 37524128 DOI: 10.1016/j.jchemneu.2023.102321] [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: 04/25/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
Prohibitin 1 (PHB1) and prohibitin 2 (PHB2) are proteins that are nearly ubiquitously expressed. They are localized in mitochondria, cytosol and cell nuclei. In the healthy CNS, they occur in neurons and non-neuronal cells (oligodendrocytes, astrocytes, microglia, and endothelial cells) and fulfill pivotal functions in brain development and aging, the regulation of brain metabolism, maintenance of structural integrity, synapse formation, aminoacidergic neurotransmission and, probably, regulation of brain action of certain hypothalamic-pituitary hormones.With regard to the diseased brain there is increasing evidence that prohibitins are prominently involved in numerous major diseases of the CNS, which are summarized and discussed in the present review (brain tumors, neurotropic viruses, Alzheimer disease, Down syndrome, Fronto-temporal and vascular dementia, dementia with Lewy bodies, Parkinson disease, Huntington disease, Multiple sclerosis, Amyotrophic lateral sclerosis, stroke, alcohol use disorder, schizophrenia and autism). Unfortunately, there is no PHB-targeted therapy available for any of these diseases.
Collapse
Affiliation(s)
- Hans-Gert Bernstein
- Department of Psychiatry, Otto-von-Guericke University, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | - Karl-Heinz Smalla
- Leibniz Institute for Neurobiology, RG Neuroplasticity, D-39118 Magdeburg, Germany; Institute for Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany, Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Gerburg Keilhoff
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University, Magdeburg, Germany
| | - Henrik Dobrowolny
- Department of Psychiatry, Otto-von-Guericke University, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Michael R Kreutz
- Leibniz Institute for Neurobiology, RG Neuroplastcity, D-39118 Magdeburg, Germany; University Medical Center Hamburg Eppendorf, Leibniz Group "Dendritic Organelles and Synaptic Function" ZMNH, Hamburg, Germany
| | - Johann Steiner
- Department of Psychiatry, Otto-von-Guericke University, Leipziger Str. 44, D-39120 Magdeburg, Germany
| |
Collapse
|
2
|
Fernandez-Abascal J, Artal-Sanz M. Prohibitins in neurodegeneration and mitochondrial homeostasis. FRONTIERS IN AGING 2022; 3:1043300. [PMID: 36404989 PMCID: PMC9674034 DOI: 10.3389/fragi.2022.1043300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
Abstract
The incidence of age-related neurodegenerative disorders has risen with the increase of life expectancy. Unfortunately, the diagnosis of such disorders is in most cases only possible when the neurodegeneration status is already advanced, and symptoms are evident. Although age-related neurodegeneration is a common phenomenon in living animals, the cellular and molecular mechanisms behind remain poorly understood. Pathways leading to neurodegeneration usually diverge from a common starting point, mitochondrial stress, which can serve as a potential target for early diagnosis and treatments. Interestingly, the evolutionarily conserved mitochondrial prohibitin (PHB) complex is a key regulator of ageing and metabolism that has been associated with neurodegenerative diseases. However, its role in neurodegeneration is still not well characterized. The PHB complex shows protective or toxic effects in different genetic and physiological contexts, while mitochondrial and cellular stress promote both up and downregulation of PHB expression. With this review we aim to shed light into the complex world of PHB’s function in neurodegeneration by putting together the latest advances in neurodegeneration and mitochondrial homeostasis associated with PHB. A better understanding of the role of PHB in neurodegeneration will add knowledge to neuron deterioration during ageing and help to identify early molecular markers of mitochondrial stress. This review will deepen our understanding of age-related neurodegeneration and provide questions to be addressed, relevant to human health and to improve the life quality of the elderly.
Collapse
Affiliation(s)
- Jesus Fernandez-Abascal
- Andalusian Centre for Developmental Biology (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, Sevilla, Spain
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, Seville, Spain
- *Correspondence: Jesus Fernandez-Abascal, ; Marta Artal-Sanz,
| | - Marta Artal-Sanz
- Andalusian Centre for Developmental Biology (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, Sevilla, Spain
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, Seville, Spain
- *Correspondence: Jesus Fernandez-Abascal, ; Marta Artal-Sanz,
| |
Collapse
|
3
|
Moore BR, Islam B, Ward S, Jackson O, Armitage R, Blackburn J, Haider S, McHugh PC. Repurposing of Tranilast for Potential Neuropathic Pain Treatment by Inhibition of Sepiapterin Reductase in the BH 4 Pathway. ACS OMEGA 2019; 4:11960-11972. [PMID: 31460307 PMCID: PMC6682008 DOI: 10.1021/acsomega.9b01228] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 06/26/2019] [Indexed: 05/08/2023]
Abstract
Tetrahydrobiopterin (BH4) is a cofactor in the production of various signaling molecules including nitric oxide, dopamine, adrenaline, and noradrenaline. BH4 levels are critical for processes associated with cardiovascular function, inflammation, mood, pain, and neurotransmission. Increasing pieces of evidence suggest that BH4 is upregulated in chronic pain. Sepiapterin reductase (SPR) catalyzes both the reversible reduction of sepiapterin to dihydrobiopterin (BH2) and 6-pyruvoyl-tetrahydrobiopterin to BH4 within the BH4 pathway. Therefore, inhibition of SPR by small molecules can be used to control BH4 production and ultimately alleviate chronic pain. Here, we have used various in silico and in vitro experiments to show that tranilast, licensed for use in bronchial asthma, can inhibit sepiapterin reduction by SPR. Docking and molecular dynamics simulations suggest that tranilast can bind to human SPR (hSPR) at the same site as sepiapterin including S157, one of the catalytic triad residues of hSPR. Colorimetric assays revealed that tranilast was nearly twice as potent as the known hSPR inhibitor, N-acetyl serotonin. Tranilast was able to inhibit hSPR activity both intracellularly and extracellularly in live cells. Triple quad mass spectrophotometry of cell lysates showed a proportional decrease of BH4 in cells treated with tranilast. Our results suggest that tranilast can act as a potent hSPR inhibitor and therefore is a valid candidate for drug repurposing in the treatment of chronic pain.
Collapse
Affiliation(s)
- Benjamin
J. R. Moore
- Centre
for Biomarker Research, School of Applied Sciences, Department of Pharmacy,
School of Applied Sciences, Innovative Physical Organic Solutions (IPOS), Department
of Chemical and Biological Sciences, and Department of Chemical Sciences,
School of Applied Sciences, University of
Huddersfield, Queensgate, Huddersfield HD1 3DH, U.K.
| | - Barira Islam
- Centre
for Biomarker Research, School of Applied Sciences, Department of Pharmacy,
School of Applied Sciences, Innovative Physical Organic Solutions (IPOS), Department
of Chemical and Biological Sciences, and Department of Chemical Sciences,
School of Applied Sciences, University of
Huddersfield, Queensgate, Huddersfield HD1 3DH, U.K.
| | - Sean Ward
- Centre
for Biomarker Research, School of Applied Sciences, Department of Pharmacy,
School of Applied Sciences, Innovative Physical Organic Solutions (IPOS), Department
of Chemical and Biological Sciences, and Department of Chemical Sciences,
School of Applied Sciences, University of
Huddersfield, Queensgate, Huddersfield HD1 3DH, U.K.
| | - Olivia Jackson
- Centre
for Biomarker Research, School of Applied Sciences, Department of Pharmacy,
School of Applied Sciences, Innovative Physical Organic Solutions (IPOS), Department
of Chemical and Biological Sciences, and Department of Chemical Sciences,
School of Applied Sciences, University of
Huddersfield, Queensgate, Huddersfield HD1 3DH, U.K.
| | - Rebecca Armitage
- Centre
for Biomarker Research, School of Applied Sciences, Department of Pharmacy,
School of Applied Sciences, Innovative Physical Organic Solutions (IPOS), Department
of Chemical and Biological Sciences, and Department of Chemical Sciences,
School of Applied Sciences, University of
Huddersfield, Queensgate, Huddersfield HD1 3DH, U.K.
| | - Jack Blackburn
- Centre
for Biomarker Research, School of Applied Sciences, Department of Pharmacy,
School of Applied Sciences, Innovative Physical Organic Solutions (IPOS), Department
of Chemical and Biological Sciences, and Department of Chemical Sciences,
School of Applied Sciences, University of
Huddersfield, Queensgate, Huddersfield HD1 3DH, U.K.
| | - Shozeb Haider
- UCL
School of Pharmacy, 29−39 Brunswick Square, London WC1N 1AX, U.K.
| | - Patrick C. McHugh
- Centre
for Biomarker Research, School of Applied Sciences, Department of Pharmacy,
School of Applied Sciences, Innovative Physical Organic Solutions (IPOS), Department
of Chemical and Biological Sciences, and Department of Chemical Sciences,
School of Applied Sciences, University of
Huddersfield, Queensgate, Huddersfield HD1 3DH, U.K.
- E-mail: . Phone: +(44) 1484 472074. Fax: +(44) 1484 472182
| |
Collapse
|
4
|
Mendez-David I, Boursier C, Domergue V, Colle R, Falissard B, Corruble E, Gardier AM, Guilloux JP, David DJ. Differential Peripheral Proteomic Biosignature of Fluoxetine Response in a Mouse Model of Anxiety/Depression. Front Cell Neurosci 2017; 11:237. [PMID: 28860968 PMCID: PMC5561647 DOI: 10.3389/fncel.2017.00237] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/26/2017] [Indexed: 01/12/2023] Open
Abstract
The incorporation of peripheral biomarkers in the treatment of major depressive disorders (MDD) could improve the efficiency of treatments and increase remission rate. Peripheral blood mononuclear cells (PBMCs) represent an attractive biological substrate allowing the identification of a drug response signature. Using a proteomic approach with high-resolution mass spectrometry, the present study aimed to identify a biosignature of antidepressant response (fluoxetine, a Selective Serotonin Reuptake Inhibitor) in PBMCs in a mouse model of anxiety/depression. Following determination of an emotionality score, using complementary behavioral analysis of anxiety/depression across three different tests (Elevated Plus Maze, Novelty Suppressed Feeding, Splash Test), we showed that a 4-week corticosterone treatment (35 μg/ml, CORT model) in C57BL/6NTac male mice induced an anxiety/depressive-like behavior. Then, chronic fluoxetine treatment (18 mg/kg/day for 28 days in the drinking water) reduced corticosterone-induced increase in emotional behavior. However, among 46 fluoxetine-treated mice, only 30 of them presented a 50% decrease in emotionality score, defining fluoxetine responders (CORT/Flx-R). To determine a peripheral biological signature of fluoxetine response, proteomic analysis was performed from PBMCs isolated from the “most” affected corticosterone/vehicle (CORT/V), corticosterone/fluoxetine responders and non-responders (CORT/Flx-NR) animals. In comparison to CORT/V, a total of 263 proteins were differently expressed after fluoxetine exposure. Expression profile of these proteins showed a strong similarity between CORT/Flx-R and CORT/Flx-NR (R = 0.827, p < 1e-7). Direct comparison of CORT/Flx-R and CORT/Flx-NR groups revealed 100 differently expressed proteins, representing a combination of markers associated either with the maintenance of animals in a refractory state, or associated with behavioral improvement. Finally, 19 proteins showed a differential direction of expression between CORT/Flx-R and CORT/Flx-NR that drove them away from the CORT-treated profile. Among them, eight upregulated proteins (RPN2, HSPA9, NPTN, AP2B1, UQCRC2, RACK-1, TOLLIP) and one downregulated protein, TLN2, were previously associated with MDD or antidepressant drug response in the literature. Future preclinical studies will be required to validate whether proteomic changes observed in PBMCs from CORT/Flx-R mice mirror biological changes in brain tissues.
Collapse
Affiliation(s)
- Indira Mendez-David
- CESP/UMR-S 1178, Université Paris-Sud, INSERM, Université Paris-SaclayChâtenay-Malabry, France
| | - Céline Boursier
- Proteomic Facility, Institut Paris Saclay d'Innovation Thérapeutique (UMS IPSIT), Université Paris-Sud, Université Paris-SaclayChâtenay-Malabry, France
| | - Valérie Domergue
- Animal Facility, Institut Paris Saclay d'Innovation Thérapeutique (UMS IPSIT), Université Paris-Sud, Université Paris-SaclayChâtenay-Malabry, France
| | - Romain Colle
- CESP/UMR 1178, Service de Psychiatrie, Faculté de Médecine, Université Paris-Sud, INSERM, Université Paris-Saclay, Hôpital BicêtreLe Kremlin Bicêtre, France
| | - Bruno Falissard
- CESP/UMR 1178, Service de Psychiatrie, Faculté de Médecine, Université Paris-Sud, INSERM, Université Paris-Saclay, Hôpital BicêtreLe Kremlin Bicêtre, France
| | - Emmanuelle Corruble
- CESP/UMR 1178, Service de Psychiatrie, Faculté de Médecine, Université Paris-Sud, INSERM, Université Paris-Saclay, Hôpital BicêtreLe Kremlin Bicêtre, France
| | - Alain M Gardier
- CESP/UMR-S 1178, Université Paris-Sud, INSERM, Université Paris-SaclayChâtenay-Malabry, France
| | - Jean-Philippe Guilloux
- CESP/UMR-S 1178, Université Paris-Sud, INSERM, Université Paris-SaclayChâtenay-Malabry, France
| | - Denis J David
- CESP/UMR-S 1178, Université Paris-Sud, INSERM, Université Paris-SaclayChâtenay-Malabry, France
| |
Collapse
|
5
|
Balasubramanian D, Deng AX, Doudney K, Hampton MB, Kennedy MA. Valproic acid exposure leads to upregulation and increased promoter histone acetylation of sepiapterin reductase in a serotonergic cell line. Neuropharmacology 2015; 99:79-88. [DOI: 10.1016/j.neuropharm.2015.06.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 05/22/2015] [Accepted: 06/26/2015] [Indexed: 01/10/2023]
|
6
|
Fluoxetine-induced regulation of heat shock protein 90 and 14-3-3ε in human embryonic carcinoma cells. Neuroreport 2015; 25:1399-404. [PMID: 25353280 DOI: 10.1097/wnr.0000000000000284] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Fluoxetine, a serotonin-selective reuptake inhibitor, exerts antidepressant and antianxiety effects on major depressive and anxiety disorders. Previous studies suggest that treatment with fluoxetine influences the expression of various proteins that are involved in proliferation, differentiation, and apoptosis in the neuronal cells of the brain. However, many aspects of the molecular pathways that modulate antidepressant action are not well understood. Here, with the aim of identifying proteins involved in antidepressant action, we examined the protein expression profile of human embryonic carcinoma (NCCIT) cells in response to fluoxetine treatment using proteomic techniques such as two-dimensional gel electrophoresis and matrix-assisted laser desorption ionization-time-of-flight mass spectrometry (MALDI-TOF MS). We found several upregulated and downregulated proteins in fluoxetine-treated NCCIT cells, and then biochemically confirmed the increased expression of heat shock protein 90 and 14-3-3ε, which play an essential role in many cellular mechanisms including cell cycle control and other signaling pathways. Our data suggest that the regulated expression of heat shock protein 90, 14-3-3ε, and other identified proteins may be associated with the therapeutic action of fluoxetine.
Collapse
|
7
|
Carboni L. The contribution of proteomic studies in humans, animal models, and after antidepressant treatments to investigate the molecular neurobiology of major depression. Proteomics Clin Appl 2015; 9:889-98. [PMID: 25488430 DOI: 10.1002/prca.201400139] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 11/03/2014] [Accepted: 12/02/2014] [Indexed: 11/07/2022]
Abstract
The neurobiological basis of major depressive disorder (MDD) is only partially understood. The proposed hypotheses postulate dysregulations of monoaminergic and other neurotransmitter pathways, impaired stress responses, insufficient neurogenetic and neurotrophic processes generating maladaptive neuroplasticity, inappropriate inflammatory and metabolic responses. Proteomic approaches can provide useful contributions to the investigation of the molecular neurobiology of MDD due to their open-ended nature. Studies performed in brain regions of MDD patients which had received antidepressant (AD) treatment showed that affected proteins mainly belonged to energy pathways, transport of molecules, signaling, and synaptic transmission. Studies performed in animal models offer the advantage of more controlled experimental conditions at the expense of potential loss in relevance. The design of proteomic investigations included experiments carried out in MDD models, in naive animals treated with ADs, and in animal models subjected to AD treatments. A comparison of results suggested an overlap of several modulated pathways between MDD patients and animal models. Examples include the regulation of energy metabolism, especially oxidative phosphorylation and glycolysis, signal transduction pathways, including calcium-calmodulin kinase II, synaptic proteins, and cytoskeletal proteins. Nevertheless, the paucity of studies performed in human brains requires additional studies to confirm the correspondence.
Collapse
Affiliation(s)
- Lucia Carboni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
8
|
Doh MS, Han DMR, Oh DH, Kim SH, Choi MR, Chai YG. Profiling of Proteins Regulated by Venlafaxine during Neural Differentiation of Human Cells. Psychiatry Investig 2015; 12:81-91. [PMID: 25670950 PMCID: PMC4310925 DOI: 10.4306/pi.2015.12.1.81] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 03/07/2014] [Accepted: 03/25/2014] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Antidepressants are known to positively influence several factors in patients with depressive disorders, resulting in increased neurogenesis and subsequent relief of depressive disorders. To study the effects of venlafaxine during neural differentiation at the cellular level, we looked at its effect on protein expression and regulation mechanisms during neural differentiation. METHODS After exposing NCCIT cell-derived EBs to venlafaxine during differentiation (1 day and 7 days), changes in protein expression were analyzed by 2-DE and MALDI-TOF MS analysis. Gene levels of proteins regulated by venlafaxine were analyzed by real-time RT-PCR. RESULTS Treatment with venlafaxine decreased expression of prolyl 4-hydroxylase (P4HB), ubiquitin-conjugating enzyme E2K (HIP2) and plastin 3 (T-plastin), and up-regulated expression of growth factor beta-3 (TGF-β3), dihydropyrimidinase-like 3 (DPYSL3), and pyruvate kinase (PKM) after differentiation for 1 and 7 days. In cells exposed to venlafaxine, the mRNA expression patterns of HIP2 and PKM, which function as negative and positive regulators of differentiation and neuronal survival, respectively, were consistent with the observed changes in protein expression. CONCLUSION Our findings may contribute to improve understanding of molecular mechanism of venlafaxine.
Collapse
Affiliation(s)
- Mi Sook Doh
- Department of Molecular and Life Sciences, Hanyang University, Ansan, Republic of Korea
| | - Dal Mu Ri Han
- Department of Molecular and Life Sciences, Hanyang University, Ansan, Republic of Korea
| | - Dong Hoon Oh
- Department of Neuropsychiatry, College of Medicine and Institute of Mental Health, Hanyang University, Seoul, Republic of Korea
| | - Seok Hyeon Kim
- Department of Neuropsychiatry, College of Medicine and Institute of Mental Health, Hanyang University, Seoul, Republic of Korea
| | - Mi Ran Choi
- Department of Molecular and Life Sciences, Hanyang University, Ansan, Republic of Korea
| | - Young Gyu Chai
- Department of Molecular and Life Sciences, Hanyang University, Ansan, Republic of Korea
| |
Collapse
|
9
|
Jodczyk S, Pearson JF, Aitchison A, Miller AL, Hampton MB, Kennedy MA. Telomere length measurement on the Roche LightCycler 480 Platform. Genet Test Mol Biomarkers 2014; 19:63-8. [PMID: 25535668 DOI: 10.1089/gtmb.2014.0208] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The average length of telomeres as measured in genomic DNA from human peripheral blood leukocytes is proving to be a potential biomarker of great interest, particularly with respect to studies of aging, specific diseases, and the effects of various stresses on overall health. AIMS The aim of this study was to establish an effective real-time quantitative polymerase chain reaction (qPCR) method for telomere length measurement on the Roche LightCycler® 480 (LC480) real-time PCR platform. METHODS Measurement of relative average telomere length was achieved by comparing products amplified from telomere-specific primers and single copy reference gene primers in a ratio (T/S). RESULTS Extensive testing led us to conclude that a modification of the original two-plate T/S assay was more compatible with this platform than the recently developed single-plate assay, and that choice of hot-start Taq polymerase and intercalating dye were critical factors. CONCLUSIONS This modified assay generates reliable measurements as judged by correlation with data derived by the telomeric restriction fragment Southern blot-based method.
Collapse
Affiliation(s)
- Sarah Jodczyk
- 1 Gene Structure and Function Laboratory, Department of Pathology, University of Otago , Christchurch, Christchurch, New Zealand
| | | | | | | | | | | |
Collapse
|
10
|
Razavi S, Jahromi M, Amirpour N, Khosravizadeh Z. Effect of sertraline on proliferation and neurogenic differentiation of human adipose-derived stem cells. Adv Biomed Res 2014; 3:97. [PMID: 24800186 PMCID: PMC4007338 DOI: 10.4103/2277-9175.129367] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 05/23/2013] [Indexed: 11/17/2022] Open
Abstract
Background: Antidepressant drugs are commonly employed for anxiety and mood disorders. Sertraline is extensively used as antidepressant in clinic. In addition, adipose tissue represents an abundant and accessible source of adult stem cells with the ability to differentiate in to multiple lineages. Therefore, human adipose-derived stem cells (hADSCs) may be useful for autologous transplantation. Materials and Methods: In the present study, we assessed the effect of antidepressant drug Sertraline on the proliferation and neurogenic differentiation of hADSCs using MTT assay and immunofluorescence technique respectively. Results: MTT assay analysis showed that 0.5 μM Sertraline significantly increased the proliferation rate of hADSCs induced cells (P < 0.05), while immunofluorescent staining indicated that Sertraline treatment during neurogenic differentiation could be decreased the percentage of glial fibrillary acidic protein and Nestin-positive cells, but did not significantly effect on the percentage of MAP2 positive cells. Conclusion: Overall, our data show that Sertraline can be promoting proliferation rate during neurogenic differentiation of hADSCs after 6 days post-induction, while Sertraline inhibits gliogenesis of induced hADSCs.
Collapse
Affiliation(s)
- Shahnaz Razavi
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maliheh Jahromi
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nushin Amirpour
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Khosravizadeh
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
11
|
Huang TL, Sung ML, Chen TY. 2D-DIGE proteome analysis on the platelet proteins of patients with major depression. Proteome Sci 2014; 12:1. [PMID: 24383611 PMCID: PMC3898786 DOI: 10.1186/1477-5956-12-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 12/04/2013] [Indexed: 01/17/2023] Open
Abstract
INTRODUCTION Platelet activation is related to the psychopathology of major depression. We attempted to search and identify protein biomarkers from the platelets of patients with major depression. High resolution two-dimensional Differential Gel Electrophoresis (2D-DIGE), the matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS), Western blot, and bioinformatic tools were applied to examine the platelet proteins of 10 patients with major depression and 10 healthy controls. RESULTS The levels of 8 proteins were significantly different between the patients with major depression in the acute phase and healthy controls. The levels of protein disulfide-isomerase A3 (PDIA3) and F-actin-capping protein subunit beta (CAPZB) were higher in patients with major depression than in healthy controls. The levels of fibrinogen beta chain (FIBB), fibrinogen gamma chain (FIBG), retinoic acid receptor beta (RARB), glutathione peroxidase 1 (GPX1), SH3 domain-containing protein 19 (SH319), and T-complex protein 1 subunit beta (TCPB) were lower in patients with major depression than in healthy controls. CONCLUSIONS Platelet provided valuable information about the pathways and processes of inflammation/immunity, oxidative stress, and neurogenesis, related to major depression.
Collapse
Affiliation(s)
- Tiao-Lai Huang
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, 123 Ta-Pei Road, Niao-Sung, Kaohsiung 833, Taiwan.
| | | | | |
Collapse
|
12
|
Kékesi KA, Juhász G, Simor A, Gulyássy P, Szegő EM, Hunyadi-Gulyás E, Darula Z, Medzihradszky KF, Palkovits M, Penke B, Czurkó A. Altered functional protein networks in the prefrontal cortex and amygdala of victims of suicide. PLoS One 2012; 7:e50532. [PMID: 23272063 PMCID: PMC3516509 DOI: 10.1371/journal.pone.0050532] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Accepted: 10/26/2012] [Indexed: 12/18/2022] Open
Abstract
Probing molecular brain mechanisms related to increased suicide risk is an important issue in biological psychiatry research. Gene expression studies on post mortem brains indicate extensive changes prior to a successful suicide attempt; however, proteomic studies are scarce. Thus, we performed a DIGE proteomic analysis of post mortem tissue samples from the prefrontal cortex and amygdala of suicide victims to identify protein changes and biomarker candidates of suicide. Among our matched spots we found 46 and 16 significant differences in the prefrontal cortex and amygdala, respectively; by using the industry standard t test and 1.3 fold change as cut off for significance. Because of the risk of false discoveries (FDR) in these data, we also made FDR adjustment by calculating the q-values for all the t tests performed and by using 0.06 and 0.4 as alpha thresholds we reduced the number of significant spots to 27 and 9 respectively. From these we identified 59 proteins in the cortex and 11 proteins in the amygdala. These proteins are related to biological functions and structures such as metabolism, the redox system, the cytoskeleton, synaptic function, and proteolysis. Thirteen of these proteins (CBR1, DPYSL2, EFHD2, FKBP4, GFAP, GLUL, HSPA8, NEFL, NEFM, PGAM1, PRDX6, SELENBP1 and VIM,) have already been suggested to be biomarkers of psychiatric disorders at protein or genome level. We also pointed out 9 proteins that changed in both the amygdala and the cortex, and from these, GFAP, INA, NEFL, NEFM and TUBA1 are interacting cytoskeletal proteins that have a functional connection to glutamate, GABA, and serotonin receptors. Moreover, ACTB, CTSD and GFAP displayed opposite changes in the two examined brain structures that might be a suitable characteristic for brain imaging studies. The opposite changes of ACTB, CTSD and GFAP in the two brain structures were validated by western blot analysis.
Collapse
Affiliation(s)
- Katalin Adrienna Kékesi
- Laboratory of Proteomics, Institute of Biology, Eötvös Loránd University, Budapest, Hungary.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Barthéléry M, Jaishankar A, Salli U, Freeman WM, Vrana KE. 2-D DIGE identification of differentially expressed heterogeneous nuclear ribonucleoproteins and transcription factors during neural differentiation of human embryonic stem cells. Proteomics Clin Appl 2012; 3:505-14. [PMID: 21136975 DOI: 10.1002/prca.200800109] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Neural stem cells (NSC) are progenitors that can give rise to all neural lineages. They are found in specific niches of fetal and adult brains and grow in vitro as non-adherent colonies, the neurospheres. These cells express the intermediate filament nestin, commonly considered an NSC marker. NSC can be derived as neurospheres from human embryonic stem cells (hESC). The mechanisms of cellular programming that hESC undergo during differentiation remain obscure. To investigate the commitment process of hESC during directed neural differentiation, we compared the nuclear proteomes of hESC and hESC-derived neurospheres. We used 2-D DIGE to conduct a quantitative comparison of hESC and NSC nuclear proteins and detected 1521 protein spots matched across three gels. Statistical analysis (ANOVA n = 3 with false discovery correction) revealed that only 2.1% of the densitometric signal was significantly changed. The ranges of average ratios varied from 1.2- to 11-fold at a statistically significant p-value <0.05. MS/MS identified 15 regulated proteins previously shown to be involved in chromatin remodeling, mRNA processing and gene expression regulation. Notably, three members of the heterogeneous nuclear ribonucleoprotein family (AUF-1, and FBP-1 and FBP-2) register a 54, 70 and 99% increased expression, highlighting them as potential markers for NSC in vitro derivation. By contrast, Cpsf-6 virtually disappears with differentiation with an 11-fold drop in NSC, highlighting this protein as a novel marker for undifferentiated ESC.
Collapse
Affiliation(s)
- Miguel Barthéléry
- Pennsylvania State University College of Medicine, Department of Pharmacology, Hummelstown, PA, USA
| | | | | | | | | |
Collapse
|
14
|
Laurindo FRM, Pescatore LA, Fernandes DDC. Protein disulfide isomerase in redox cell signaling and homeostasis. Free Radic Biol Med 2012; 52:1954-69. [PMID: 22401853 DOI: 10.1016/j.freeradbiomed.2012.02.037] [Citation(s) in RCA: 182] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 02/23/2012] [Accepted: 02/24/2012] [Indexed: 12/16/2022]
Abstract
Thiol proteins may potentially act as redox signaling adaptor proteins, adjusting reactive oxygen species intermediates to specific signals and redox signals to cell homeostasis. In this review, we discuss redox effects of protein disulfide isomerase (PDI), a thioredoxin superfamily oxidoreductase from the endoplasmic reticulum (ER). Abundantly expressed PDI displays ubiquity, interactions with redox and nonredox proteins, versatile effects, and several posttranslational modifications. The PDI family contains >20 members with at least some apparent complementary actions. PDI has oxidoreductase, isomerase, and chaperone effects, the last not directly dependent on its thiols. PDI is a converging hub for pathways of disulfide bond introduction into ER-processed proteins, via hydrogen peroxide-generating mechanisms involving the oxidase Ero1α, as well as hydrogen peroxide-consuming reactions involving peroxiredoxin IV and the novel peroxidases Gpx7/8. PDI is a candidate pathway for coupling ER stress to oxidant generation. Emerging information suggests a convergence between PDI and Nox family NADPH oxidases. PDI silencing prevents Nox responses to angiotensin II and inhibits Akt phosphorylation in vascular cells and parasite phagocytosis in macrophages. PDI overexpression spontaneously enhances Nox activation and expression. In neutrophils, PDI redox-dependently associates with p47phox and supports the respiratory burst. At the cell surface, PDI exerts transnitrosation, thiol reductase, and apparent isomerase activities toward targets including adhesion and matrix proteins and proteases. Such effects mediate redox-dependent adhesion, coagulation/thrombosis, immune functions, and virus internalization. The route of PDI externalization remains elusive. Such multiple redox effects of PDI may contribute to its conspicuous expression and functional role in disease, rendering PDI family members putative redox cell signaling adaptors.
Collapse
Affiliation(s)
- Francisco R M Laurindo
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, 05403-000 São Paulo, Brazil.
| | | | | |
Collapse
|
15
|
Webhofer C, Gormanns P, Tolstikov V, Zieglgänsberger W, Sillaber I, Holsboer F, Turck CW. Metabolite profiling of antidepressant drug action reveals novel drug targets beyond monoamine elevation. Transl Psychiatry 2011; 1:e58. [PMID: 22832350 PMCID: PMC3309495 DOI: 10.1038/tp.2011.56] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 10/25/2011] [Accepted: 11/01/2011] [Indexed: 11/09/2022] Open
Abstract
Currently used antidepressants elevate monoamine levels in the synaptic cleft. There is good reason to assume that this is not the only source for antidepressant therapeutic activities and that secondary downstream effects may be relevant for alleviating symptoms of depression. We attempted to elucidate affected biochemical pathways downstream of monoamine reuptake inhibition by interrogating metabolomic profiles in DBA/2Ola mice after chronic paroxetine treatment. Metabolomic changes were investigated using gas chromatography-mass spectrometry profiling and group differences were analyzed by univariate and multivariate statistics. Pathways affected by antidepressant treatment were related to energy metabolism, amino acid metabolism and hormone signaling. The identified pathways reveal further antidepressant therapeutic action and represent targets for drug development efforts. A comparison of the central nervous system with blood plasma metabolite alterations identified GABA, galactose-6-phosphate and leucine as biomarker candidates for assessment of antidepressant treatment effects in the periphery.
Collapse
Affiliation(s)
- C Webhofer
- Max Planck Institute of Psychiatry, Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - P Gormanns
- Max Planck Institute of Psychiatry, Munich, Germany
| | | | - W Zieglgänsberger
- Max Planck Institute of Psychiatry, Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - I Sillaber
- Max Planck Institute of Psychiatry, Munich, Germany
- Phenoquest AG, Martinsried, Germany
| | - F Holsboer
- Max Planck Institute of Psychiatry, Munich, Germany
| | - C W Turck
- Max Planck Institute of Psychiatry, Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| |
Collapse
|
16
|
McHugh PC. The tetrahydrobiopterin pathway: a novel target for the treatment of depression. Pharmacogenomics 2011; 12:1625-7. [PMID: 22118047 DOI: 10.2217/pgs.11.138] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
17
|
Piubelli C, Gruber S, El Khoury A, Mathé AA, Domenici E, Carboni L. Nortriptyline influences protein pathways involved in carbohydrate metabolism and actin-related processes in a rat gene-environment model of depression. Eur Neuropsychopharmacol 2011; 21:545-62. [PMID: 21168998 DOI: 10.1016/j.euroneuro.2010.11.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Revised: 09/24/2010] [Accepted: 11/09/2010] [Indexed: 01/21/2023]
Abstract
Although most available antidepressants increase monoaminergic neurotransmission, their therapeutic efficacy is likely mediated by longer-term molecular adaptations. To investigate the molecular changes induced by chronic antidepressant treatment we analysed proteomic changes in rat pre-frontal/frontal cortex and hippocampus after nortriptyline (NT) administration. A wide-scale analysis of protein expression was performed on the Flinders Sensitive Line (FSL), a genetically-selected rat model of depression, and the control Flinders Resistant Line (FRL). The effect of NT treatment was examined in a gene-environment interaction model, applying maternal separation (MS) to both strains. In the forced swim test, FSL rats were significantly more immobile than FRL animals, whereas NT treatment reduced immobility time. MS alone did not modify immobility time, but it impaired the response to NT in the FSL strain. In the proteomic analysis, in FSL rats NT treatment chiefly modulated cytoskeleton proteins and carbohydrate metabolism. In the FRL strain, changes influenced protein polymerization and intracellular transport. After MS, NT treatment mainly affected proteins in nucleotide metabolism in FSL rats and synaptic transmission and neurite morphogenesis pathways in FRL rats. When the effects of NT treatment and MS were compared between strains, carbohydrate metabolic pathways were predominantly modulated.
Collapse
|
18
|
Escitalopram modulates neuron-remodelling proteins in a rat gene-environment interaction model of depression as revealed by proteomics. Part I: genetic background. Int J Neuropsychopharmacol 2011; 14:796-833. [PMID: 21054914 DOI: 10.1017/s1461145710001318] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The wide-scale analysis of protein expression provides a powerful strategy for the molecular exploration of complex pathophysiological mechanisms, such as the response to antidepressants. Using a 2D proteomic approach we investigated the Flinders Sensitive Line (FSL), a genetically selected rat model of depression, and the control Flinders Resistant Line (FRL). To evaluate gene-environment interactions, FSL and FRL pups were separated from their mothers for 3 h (maternal separation, MS), as early-life trauma is considered an important antecedent of depression. All groups were treated with either escitalopram (Esc) admixed to food (25 mg/kg.d) or vehicle for 1 month. At the week 3, forced swim tests were performed. Protein extracts from prefrontal/frontal cortex and hippocampus were separated by 2D electrophoresis. Proteins displaying statistically significant differences in expression levels were identified by mass spectrometry. Immobility time values in the forced swim test were higher in FSL rats and reduced by antidepressant treatment. Moreover, the Esc-induced reduction in immobility time was not detected in MS rats. The impact of genetic background in response to Esc was specifically investigated here. Bioinformatics analyses highlighted gene ontology terms showing tighter associations with the modulated proteins. Esc modulated protein belonging to cytoskeleton organization in FSL; carbohydrate metabolism and intracellular transport in FRL. Proteins differently modulated in the two strains after MS and Esc play a role in cytoskeleton organization, vesicle-mediated transport, apoptosis regulation and macromolecule catabolism. These findings suggest pathways involved in neuronal remodelling as molecular correlates of response to antidepressants in a model of vulnerability.
Collapse
|
19
|
McHugh PC, Joyce PR, Deng X, Kennedy MA. A polymorphism of the GTP-cyclohydrolase I feedback regulator gene alters transcriptional activity and may affect response to SSRI antidepressants. THE PHARMACOGENOMICS JOURNAL 2011; 11:207-13. [PMID: 20351752 DOI: 10.1038/tpj.2010.23] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Revised: 01/03/2010] [Accepted: 02/28/2010] [Indexed: 11/09/2022]
Abstract
Tetrahydrobiopterin (BH(4)) is an essential cofactor for synthesis of many neurotransmitters including serotonin. In serotonergic neurons, BH(4) is tightly regulated by GTP-cyclohydrolase I feedback regulator (GFRP). Given the pivotal role of the serotonergic system in mood disorders and selective serotonin reuptake inhibitors (SSRIs) antidepressant function, we tested the hypothesis that GFRP gene (GCHFR) variants would modify response to antidepressants in subjects with major depression. Two single nucleotide polymorphisms (rs7164342 and rs7163862) in the GCHFR promoter were identified and occurred as two haplotypes (GA or TT). A multiple regression analysis revealed that homozygous individuals for the TT haplotype were less likely to respond to the SSRI fluoxetine than to the tricyclic antidepressant nortriptyline (P = 0.037). Moreover, the TT haplotype showed a reduced transcription rate in luciferase reporter gene assays, which may impact on BH(4)-mediated neurotransmitter production, thus suggesting a biological process through which GCHFR promoter variants might influence antidepressant response.
Collapse
Affiliation(s)
- P C McHugh
- Department of Pathology, University of Otago, Christchurch, New Zealand.
| | | | | | | |
Collapse
|
20
|
Scaini G, Maggi DD, De-Nês BT, Gonçalves CL, Ferreira GK, Teodorak BP, Bez GD, Ferreira GC, Schuck PF, Quevedo J, Streck EL. Activity of mitochondrial respiratory chain is increased by chronic administration of antidepressants. Acta Neuropsychiatr 2011; 23:112-8. [PMID: 26952897 DOI: 10.1111/j.1601-5215.2011.00548.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Depressive disorders, including major depression, are serious and disabling for affected patients. Although the neurobiological understanding of major depressive disorder focuses mainly on the monoamine hypothesis, the exact pathophysiology of depression is not fully understood. METHODS Animals received daily intra-peritoneal injections of paroxetine (10 mg/kg), nortriptyline (15 mg/kg) or venlafaxine (10 mg/kg) in 1.0 ml/kg volume for 15 days. Twelve hours after the last injection, the rats were killed by decapitation, where the brain was removed and homogenised. The activities of mitochondrial respiratory chain complexes in different brain structures were measured. RESULTS We first verified that chronic administration of paroxetine increased complex I activity in prefrontal cortex, hippocampus, striatum and cerebral cortex. In addition, complex II activity was increased by the same drug in hippocampus, striatum and cerebral cortex and complex IV activity in prefrontal cortex. Furthermore, chronic administration of nortriptyline increased complex II activity in hippocampus and striatum and complex IV activity in prefrontal cortex, striatum and cerebral cortex. Finally, chronic administration of venlafaxine increased complex II activity in hippocampus, striatum and cerebral cortex and complex IV activity in prefrontal cortex. CONCLUSION On the basis of the present findings, it is tempting to speculate that an increase in brain energy metabolism by the antidepressant paroxetine, nortriptyline and venlafaxine could play a role in the mechanism of action of these drugs. These data corroborate with other studies suggesting that some antidepressants modulate brain energy metabolism.
Collapse
Affiliation(s)
- Giselli Scaini
- Laboratório de Fisiopatologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Débora D Maggi
- Laboratório de Fisiopatologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Bruna T De-Nês
- Laboratório de Fisiopatologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Cinara L Gonçalves
- Laboratório de Fisiopatologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Gabriela K Ferreira
- Laboratório de Fisiopatologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Brena P Teodorak
- Laboratório de Fisiopatologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Gisele D Bez
- Laboratório de Fisiopatologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Gustavo C Ferreira
- Programa de Pós-graduação em Ciências da Saúde, Universidade do Sul de Santa Catarina, Tubarão, SC, Brazil
| | - Patricia F Schuck
- Laboratório de Fisiopatologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - João Quevedo
- Instituto Nacional de Ciência e Tecnologia Translacional em Medicina
| | - Emilio L Streck
- Laboratório de Fisiopatologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| |
Collapse
|
21
|
McHugh PC, Wright JA, Brown DR. Transcriptional regulation of the beta-synuclein 5'-promoter metal response element by metal transcription factor-1. PLoS One 2011; 6:e17354. [PMID: 21386983 PMCID: PMC3046239 DOI: 10.1371/journal.pone.0017354] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Accepted: 01/29/2011] [Indexed: 12/24/2022] Open
Abstract
The progression of many human neurodegenerative disorders is associated with an accumulation of alpha-synuclein. Alpha-synuclein belongs to the homologous synuclein family, which includes beta-synuclein. It has been proposed that beta-synuclein may be a natural regulator of alpha-synuclein. Therefore controlling beta-synuclein expression may control the accumulation of alpha-synuclein and ultimately prevent disease progression. The regulation of synucleins is poorly understood. We investigated the transcriptional regulation of beta-synuclein, with the aim of identifying molecules that differentially control beta-synuclein expression levels. To investigate transcriptional regulation of beta-synuclein, we used reporter gene assays and bioinformatics. We identified a region -1.1/-0.6 kb upstream of the beta-synuclein translational start site to be a key regulatory region of beta-synuclein 5'-promoter activity in human dopaminergic cells (SH-SY5Y). Within this key promoter region we identified a metal response element pertaining to a putative Metal Transcription Factor-1 (MTF-1) binding site. We demonstrated that MTF-1 binds to this 5'-promoter region using EMSA analysis. Moreover, we showed that MTF-1 differentially regulates beta-synuclein promoter binding site, as well as beta-synuclein mRNA and protein expression. This effect of MTF-1 on expression was found to be specific to beta-synuclein when compared to alpha-synuclein. Understanding the regulation of synucleins and how they interact may point to molecular targets that could be manipulated for therapeutic benefit. In this study we showed that MTF-1 differentially controls the expression of beta-synuclein when compared to its homolog alpha-synuclein. This could potentially provide a novel targets or pathways for therapeutic intervention and/or treatment of synucleinopathies.
Collapse
Affiliation(s)
- Patrick C. McHugh
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath, United Kingdom
| | - Josephine A. Wright
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath, United Kingdom
| | - David R. Brown
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath, United Kingdom
| |
Collapse
|
22
|
McHugh PC, Rogers GR, Glubb DM, Joyce PR, Kennedy MA. Proteomic analysis of rat hippocampus exposed to the antidepressant paroxetine. J Psychopharmacol 2010; 24:1243-51. [PMID: 19346281 DOI: 10.1177/0269881109102786] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Antidepressant drugs can exert significant effects on the mood of a patient suffering major depression and other disorders. These drugs generally have pharmacological actions on the uptake or metabolism of the neurotransmitters serotonin, noradrenaline and, to a lesser extent, dopamine. However, there are many aspects of antidepressant action we do not understand. We have applied proteomic analysis in a rat hippocampal model in an attempt to identify relevant molecules that operate in pathways functionally relevant to antidepressant action. Rats were administered either 5 mg/kg daily of the antidepressant paroxetine or vehicle for 12 days, then hippocampal protein was recovered and resolved by 2-D gel electrophoresis. After antidepressant exposure, we observed increased expression or modification of cytochrome c oxidase, subunit Va, cyclin-dependent kinase inhibitor 2A interacting protein, dynein, axonemal, heavy polypeptide 3 and RHO GDP-dissociation inhibitor alpha. Decreased expression or modification was observed for complexin 1 (CPLX1), alpha-synuclein, parvalbumin, ribosomal protein large P2, prohibitin, nerve growth factor, beta subunit (NGFB), peroxiredoxin 6 (PRDX6), 1-acylglycerol-3-phosphate O-acyltransferase 2_predicted, cystatin B (CYTB) and lysosomal membrane glycoprotein 1. CPLX1, the most strongly regulated protein observed, mediates the fusion of cellular transport vesicles with their target membranes and has been implicated in the pathophysiology of mood disorders, as well as antidepressant action. CPLX1 and the other proteins identified may represent links into molecular processes of importance to mood dysregulation and control, and their respective genes may represent novel candidates for studies of antidepressant pharmacogenetics.
Collapse
Affiliation(s)
- P C McHugh
- Department of Pathology, University of Otago, Christchurch, New Zealand.
| | | | | | | | | |
Collapse
|
23
|
Polymorphisms of sepiapterin reductase gene alter promoter activity and may influence risk of bipolar disorder. Pharmacogenet Genomics 2009; 19:330-7. [PMID: 19415819 DOI: 10.1097/fpc.0b013e328328f82c] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVES In a previous investigation, we observed altered expression of sepiapterin reductase (SPR) in cultured neural cells chronically exposed to paroxetine. SPR is an enzyme, which catalyzes the final step in the synthesis of tetrahydrobiopterin (BH4). BH4 is an essential cofactor for synthesis of many neurotransmitters including serotonin. Given the pivotal role of SPR in neurotransmitter production, we sought to test the hypothesis that SPR would influence susceptibility to mood disorders and patient response to antidepressants. METHODS We tested for association of SPR promoter polymorphisms with antidepressant response in a well-characterized triad cohort of mood disorders. We evaluated the functional effect of these variants using the Dual-Luciferase Reporter Gene Assay System in two independent cell lines. RESULTS Two promoter single nucleotide polymorphisms (rs1876487 and rs2421095) in SPR were identified that occurred in three distinct haplotypes. We found a statistically significant association of haplotype pair 2,3 with bipolar I disorder [odds ratio: 5.47; 95% confidence interval: (1.68-17.88); P<0.005] and the personality measure self-transcendence (P = 0.020). Moreover, we found preliminary evidence that individuals with haplotype pair 2,3 responded better to the treatment with selective serotonin reuptake inhibitors. Reporter gene assays revealed a 1.4-fold to 1.6-fold decrease in the transcription rate of the two less common haplotypes (2 and 3) compared with haplotype 1, in the two cell lines investigated. CONCLUSION This reduced transcription rate for SPR promoter haplotypes 2 and 3 may impact on BH4-mediated neurotransmitter production, thus suggesting a biological process through which SPR gene variants might influence antidepressant response and susceptibility to bipolar disorder.
Collapse
|
24
|
Fania C, Anastasia L, Vasso M, Papini N, Capitanio D, Venerando B, Gelfi C. Proteomic signature of reversine-treated murine fibroblasts by 2-D difference gel electrophoresis and MS: Possible associations with cell signalling networks. Electrophoresis 2009; 30:2193-206. [DOI: 10.1002/elps.200800800] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
25
|
McHugh PC, Rogers GR, Glubb DM, Allington MD, Hughes M, Joyce PR, Kennedy MA. Downregulation of Ccnd1 and Hes6 in rat hippocampus after chronic exposure to the antidepressant paroxetine. Acta Neuropsychiatr 2008; 20:307-13. [PMID: 25384412 DOI: 10.1111/j.1601-5215.2008.00334.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE The mechanism of action of antidepressant drugs is not fully understood. Application of genomic methods enables the identification of biochemical pathways that are regulated by antidepressants, and this may provide novel clues to the molecular and cellular actions of these drugs. The present study examined gene expression profiles in the hippocampus of rats exposed to chronic antidepressant treatment. METHODS Animals were treated for 12 days with the selective serotonin reuptake inhibitor paroxetine; then, hippocampal ribonucleic acid was recovered, and changes in gene expression were assessed by microarray analysis. RESULTS A total of 160 genes that showed differential expression after paroxetine exposure were identified. Using functional relevance and observed fold change as selection criteria, the expression changes in a subset of these genes were confirmed by quantitative polymerase chain reaction. CONCLUSION Of this subset, only two genes, cyclin D1 (Ccnd1) and hairy and enhancer of split 6 (Hes6), showed robust and consistent changes in expression. Both genes were downregulated by paroxetine, and both have been previously implicated in neurogenesis. Further investigation of these two genes may provide new insight into the mechanism of action of antidepressants.
Collapse
Affiliation(s)
- Patrick C McHugh
- 1Department of Pathology, University of Otago, Christchurch, Christchurch, New Zealand
| | - Geraldine R Rogers
- 1Department of Pathology, University of Otago, Christchurch, Christchurch, New Zealand
| | - Dylan M Glubb
- 1Department of Pathology, University of Otago, Christchurch, Christchurch, New Zealand
| | - Melanie D Allington
- 1Department of Pathology, University of Otago, Christchurch, Christchurch, New Zealand
| | - Mark Hughes
- 2Genetics Factors, Riccarton, Christchurch, New Zealand
| | - Peter R Joyce
- 3Department of Psychological Medicine, University of Otago, Christchurch, Christchurch, New Zealand
| | - Martin A Kennedy
- 1Department of Pathology, University of Otago, Christchurch, Christchurch, New Zealand
| |
Collapse
|
26
|
Kronsbein HC, Jastorff AM, Maccarrone G, Stalla G, Wurst W, Holsboer F, Turck CW, Deussing JM. CRHR1-dependent effects on protein expression and posttranslational modification in AtT-20 cells. Mol Cell Endocrinol 2008; 292:1-10. [PMID: 18582531 DOI: 10.1016/j.mce.2008.05.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2008] [Revised: 05/26/2008] [Accepted: 05/27/2008] [Indexed: 01/11/2023]
Abstract
Corticotropin-releasing hormone (CRH) plays a major role in coordinating the organism's stress response, including the activity of the hypothalamic-pituitary-adrenocortical axis. The molecular underpinnings of CRH-dependent signal transduction mechanisms in the anterior pituitary have not yet been revealed in detail. In order to dissect the signal transduction cascades activated by CRH receptor type 1, a comparative proteome approach was performed in vitro utilizing murine corticotroph AtT-20 cells. Alterations in protein expression and posttranslational modification in response to CRH stimulation were studied by 2D gel electrophoresis. Selected candidates were analyzed by immunoblotting and quantitative real-time PCR. The differential analyses revealed proteins regulated or modified related to diverse cellular processes. Amongst others we identified alterations in PRKAR1A, the regulatory subunit of protein kinase A; in PGK1 and PGAM1, key regulators of glycolysis; and in proteins involved in proteasome-mediated proteolysis, PSMC2 and PSMA3. These results offer novel entry points to molecular mechanisms underlying stress responses elicited via the hypothalamic-pituitary-adrenocortical axis.
Collapse
Affiliation(s)
- Helena C Kronsbein
- Max Planck Institute of Psychiatry, Kraepelinstrasse 2-10, D-80804 Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|