1
|
Avila JA, Kiprowska M, Jean-Louis T, Rockwell P, Figueiredo-Pereira ME, Serrano PA. PACAP27 mitigates an age-dependent hippocampal vulnerability to PGJ2-induced spatial learning deficits and neuroinflammation in mice. Brain Behav 2020; 10:e01465. [PMID: 31769222 PMCID: PMC6955932 DOI: 10.1002/brb3.1465] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/20/2019] [Accepted: 10/13/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Inflammation in the brain is mediated by the cyclooxygenase pathway, which leads to the production of prostaglandins. Prostaglandin (PG) D2, the most abundant PG in the brain, increases under pathological conditions and is spontaneously metabolized to PGJ2. PGJ2 is highly neurotoxic, with the potential to transition neuroinflammation into a chronic state and contribute to neurodegeneration as seen in many neurological diseases. Conversely, PACAP27 is a lipophilic peptide that raises intracellular cAMP and is an anti-inflammatory agent. The aim of our study was to investigate the therapeutic potential of PACAP27 to counter the behavioral and neurotoxic effects of PGJ2 observed in aged subjects. METHODS PGJ2 was injected bilaterally into the hippocampal CA1 region of 53-week-old and 12-week-old C57BL/6N male mice, once per week over 3 weeks (three total infusions) and included co-infusions of PACAP27 within respective treatment groups. Our behavioral assessments looked at spatial learning and memory performance on the 8-arm radial maze, followed by histological analyses of fixed hippocampal tissue using Fluoro-Jade C and fluorescent immunohistochemistry focused on IBA-1 microglia. RESULTS Aged mice treated with PGJ2 exhibited spatial learning and long-term memory deficits, as well as neurodegeneration in CA3 pyramidal neurons. Aged mice that received co-infusions of PACAP27 exhibited remediated learning and memory performance and decreased neurodegeneration in CA3 pyramidal neurons. Moreover, microglial activation in the CA3 region was also reduced in aged mice cotreated with PACAP27. CONCLUSIONS Our data show that PGJ2 can produce a retrograde spread of damage not observed in PGJ2-treated young mice, leading to age-dependent neurodegeneration of hippocampal neurons producing learning and memory deficits. PACAP27 can remediate the behavioral and neurodegenerative effects that PGJ2 produces in aged subjects. Targeting specific neurotoxic prostaglandins, such as PGJ2, offers great promise as a new therapeutic strategy downstream of cyclooxygenases, to combat the neuronal deficits induced by chronic inflammation.
Collapse
Affiliation(s)
- Jorge A Avila
- Department of Psychology, Hunter College, City University of New York, New York, NY, USA.,The Graduate Center of CUNY, New York, NY, USA
| | - Magdalena Kiprowska
- The Graduate Center of CUNY, New York, NY, USA.,Department of Biological Sciences, Hunter College, City University of New York, New York, NY, USA
| | - Teneka Jean-Louis
- The Graduate Center of CUNY, New York, NY, USA.,Department of Biological Sciences, Hunter College, City University of New York, New York, NY, USA
| | - Patricia Rockwell
- The Graduate Center of CUNY, New York, NY, USA.,Department of Biological Sciences, Hunter College, City University of New York, New York, NY, USA
| | - Maria E Figueiredo-Pereira
- The Graduate Center of CUNY, New York, NY, USA.,Department of Biological Sciences, Hunter College, City University of New York, New York, NY, USA
| | - Peter A Serrano
- Department of Psychology, Hunter College, City University of New York, New York, NY, USA.,The Graduate Center of CUNY, New York, NY, USA
| |
Collapse
|
2
|
Waschek JA, Cohen JR, Chi GC, Proszynski TJ, Niewiadomski P. PACAP Promotes Matrix-Driven Adhesion of Cultured Adult Murine Neural Progenitors. ASN Neuro 2017; 9:1759091417708720. [PMID: 28523979 PMCID: PMC5439654 DOI: 10.1177/1759091417708720] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
New neurons are born throughout the life of mammals in germinal zones of the brain known as neurogenic niches: the subventricular zone of the lateral ventricles and the subgranular zone of the dentate gyrus of the hippocampus. These niches contain a subpopulation of cells known as adult neural progenitor cells (aNPCs), which self-renew and give rise to new neurons and glia. aNPCs are regulated by many factors present in the niche, including the extracellular matrix (ECM). We show that the neuropeptide PACAP (pituitary adenylate cyclase-activating polypeptide) affects subventricular zone-derived aNPCs by increasing their surface adhesion. Gene array and reconstitution assays indicate that this effect can be attributed to the regulation of ECM components and ECM-modifying enzymes in aNPCs by PACAP. Our work suggests that PACAP regulates a bidirectional interaction between the aNPCs and their niche: PACAP modifies ECM production and remodeling, in turn the ECM regulates progenitor cell adherence. We speculate that PACAP may in this manner help restrict adult neural progenitors to the stem cell niche in vivo, with potential significance for aNPC function in physiological and pathological states.
Collapse
Affiliation(s)
- James A Waschek
- 1 Intellectual Development and Disabilities Research Center, The David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Joseph R Cohen
- 1 Intellectual Development and Disabilities Research Center, The David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Gloria C Chi
- 1 Intellectual Development and Disabilities Research Center, The David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Tomasz J Proszynski
- 2 Department of Cell Biology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Pawel Niewiadomski
- 1 Intellectual Development and Disabilities Research Center, The David Geffen School of Medicine, University of California, Los Angeles, CA, USA.,2 Department of Cell Biology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.,3 Centre of New Technologies, University of Warsaw, Warsaw, Poland
| |
Collapse
|
3
|
Takuma K, Ago Y, Hasebe S, Nakazawa T, Hashimoto H, Matsuda T. Regulation of emotional behaviors by juvenile environmental factors. Nihon Yakurigaku Zasshi 2017; 149:76-78. [PMID: 28154301 DOI: 10.1254/fpj.149.76] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
4
|
PACAP Protects the Adolescent and Adult Mice Brain from Ethanol Toxicity and Modulates Distinct Sets of Genes Regulating Similar Networks. Mol Neurobiol 2016; 54:7534-7548. [PMID: 27826748 DOI: 10.1007/s12035-016-0204-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 10/11/2016] [Indexed: 12/30/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a 38-amino acid neuropeptide which has been shown to exert various neuroprotective actions in vitro and in vivo; however, the ability of endogenous PACAP to prevent cell death in vivo remains to be elucidated. To explore the capacity of endogenous PACAP to prevent ethanol toxicity, adolescent and adult PACAP knockout (KO) mice were injected with ethanol in a binge drinking-like manner. Biochemical analyses revealed that ethanol administration induced an increase in the production of reactive oxygen species and the activity of caspase-3 in PACAP KO mice in an age-independent manner. In order to characterize the mechanisms underlying the sensitivity of PACAP KO mice, a whole-genome microarray analysis was performed to compare gene regulations induced by ethanol in adolescent and adult wild-type and PACAP KO mice. Gene expression substantially differed between adolescent and adult wild-type mice, suggesting distinct effects of ethanol according to the state of brain maturation. Interestingly, in adolescent and adult PACAP KO mice, the set of genes regulated were also markedly different but seemed to inhibit some similar regulatory network processes associated in particular with DNA repair and cell cycle. These data imply that ethanol induces serious DNA damages and cell cycle alteration in PACAP KO mice. This hypothesis, based on the transcriptomic data, could be confirmed by functional studies which showed that cell proliferation decreased in adolescent and adult PACAP KO mice treated with ethanol but recovered after a 30-day withdrawal period. These data, obtained with PACAP KO animals, demonstrate that endogenous PACAP protects the brain of adolescent and adult mice from alcohol toxicity and modulates distinct sets of genes according to the maturation status of the brain.
Collapse
|
5
|
Mansouri S, Lietzau G, Lundberg M, Nathanson D, Nyström T, Patrone C. Pituitary Adenlylate Cyclase Activating Peptide Protects Adult Neural Stem Cells from a Hypoglycaemic milieu. PLoS One 2016; 11:e0156867. [PMID: 27305000 PMCID: PMC4909203 DOI: 10.1371/journal.pone.0156867] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 05/19/2016] [Indexed: 12/25/2022] Open
Abstract
Hypoglycaemia is a common side-effect of glucose-lowering therapies for type-2 diabetic patients, which may cause cognitive/neurological impairment. Although the effects of hypoglycaemia in the brain have been extensively studied in neurons, how hypoglycaemia impacts the viability of adult neural stem cells (NSCs) has been poorly investigated. In addition, the cellular and molecular mechanisms of how hypoglycaemia regulates NSCs survival have not been characterized. Recent work others and us have shown that the pituitary adenylate cyclase-activating polypeptide (PACAP) and the glucagon-like peptide-1 receptor (GLP-1R) agonist Exendin-4 stimulate NSCs survival against glucolipoapoptosis. The aim of this study was to establish an in vitro system where to study the effects of hypoglycaemia on NSC survival. Furthermore, we determine the potential role of PACAP and Exendin-4 in counteracting the effect of hypoglycaemia. A hypoglycaemic in vitro milieu was mimicked by exposing subventricular zone-derived NSC to low levels of glucose. Moreover, we studied the potential involvement of apoptosis and endoplasmic reticulum stress by quantifying protein levels of Bcl-2, cleaved caspase-3 and mRNA levels of CHOP. We show that PACAP via PAC-1 receptor and PKA activation counteracts impaired NSC viability induced by hypoglycaemia. The protective effect induced by PACAP correlated with endoplasmic reticulum stress, Exendin-4 was ineffective. The results show that hypoglycaemia decreases NSC viability and that this effect can be substantially counteracted by PACAP via PAC-1 receptor activation. The data supports a potential therapeutic role of PAC-1 receptor agonists for the treatment of neurological complications, based on neurogenesis impairment by hypoglycaemia.
Collapse
Affiliation(s)
- Shiva Mansouri
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| | - Grazyna Lietzau
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mathias Lundberg
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - David Nathanson
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Nyström
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Cesare Patrone
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
6
|
Matsuda T. Psychopharmacological Studies in Mice. YAKUGAKU ZASSHI 2016; 136:737-50. [PMID: 27150930 DOI: 10.1248/yakushi.15-00282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Since 1998, when the laboratory of Medicinal Pharmacology was established in the Graduate School of Pharmaceutical Sciences, Osaka University, I have been interested in psychopharmacological research topics. During this period, we identified a number of novel regulatory mechanisms that control the prefrontal dopamine system through functional interaction between serotonin1A and dopamine D2 receptors or between serotonin1A and σ1 receptors. Our findings suggest that strategies that enhance the prefrontal dopamine system may have therapeutic potential in the treatment of psychiatric disorders. We also found that environmental factors during development strongly impact the psychological state in adulthood. Furthermore, we clarified the pharmacological profiles of the acetylcholinesterase inhibitors donepezil, galantamine, and rivastigmine, providing novel insights into their mechanisms of action. Finally, we developed the female encounter test, a novel method for evaluating motivation in mice. This simple method should help advance future psychopharmacological research. In this review, we summarize the major findings obtained from our recent studies in mice.
Collapse
Affiliation(s)
- Toshio Matsuda
- Laboratory of Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University
| |
Collapse
|
7
|
Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) Is Involved in Adult Mouse Hippocampal Neurogenesis After Stroke. J Mol Neurosci 2016; 59:270-9. [PMID: 26910758 DOI: 10.1007/s12031-016-0731-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 02/15/2016] [Indexed: 12/30/2022]
Abstract
In the subgranular zone (SGZ) of the hippocampus, neurogenesis persists throughout life and is upregulated following ischemia. Accumulating evidence suggests that enhanced neurogenesis stimulated by ischemic injury contributes to recovery after stroke. However, the mechanisms underlying the upregulation of neurogenesis are unclear. We have demonstrated that a neuropeptide, pituitary adenylate cyclase-activating polypeptide (PACAP), exerts a wide range of effects on neural stem cells (NSCs) during neural development. Here, we examined the effects of endogenous and exogenous PACAP in adult NSCs of the SGZ. Immunostaining showed expression of the PACAP receptor PAC1R in nestin-positive NSCs of adult naive mice. PACAP injection into the lateral ventricle increased bromodeoxyuridine (BrdU)-positive proliferative cells in the SGZ. These data suggest that PACAP promoted the proliferation of NSCs. In global ischemia model mice, the number of BrdU-positive cells was increased in wild-type mice but not in PACAP heterozygous knockout mice. The BrdU-positive cells that increased in number after ischemia were immunopositive for SOX2, a marker of NSCs, and differentiated into NeuN-positive mature neurons at 4 weeks after ischemia. These findings suggest that PACAP contributes to the proliferation of NSCs and may be associated with recovery after brain injury.
Collapse
|
8
|
PACAP enhances axon outgrowth in cultured hippocampal neurons to a comparable extent as BDNF. PLoS One 2015; 10:e0120526. [PMID: 25807538 PMCID: PMC4373823 DOI: 10.1371/journal.pone.0120526] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Accepted: 01/26/2015] [Indexed: 11/19/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) exerts neurotrophic activities including modulation of synaptic plasticity and memory, hippocampal neurogenesis, and neuroprotection, most of which are shared with brain-derived neurotrophic factor (BDNF). Therefore, the aim of this study was to compare morphological effects of PACAP and BDNF on primary cultured hippocampal neurons. At days in vitro (DIV) 3, PACAP increased neurite length and number to similar levels by BDNF, but vasoactive intestinal polypeptide showed much lower effects. In addition, PACAP increased axon, but not dendrite, length, and soma size at DIV 3 similarly to BDNF. The PACAP antagonist PACAP6–38 completely blocked the PACAP-induced increase in axon, but not dendrite, length. Interestingly, the BDNF-induced increase in axon length was also inhibited by PACAP6–38, suggesting a mechanism involving PACAP signaling. K252a, a TrkB receptor inhibitor, inhibited axon outgrowth induced by PACAP and BDNF without affecting dendrite length. These results indicate that in primary cultured hippocampal neurons, PACAP shows morphological actions via its cognate receptor PAC1, stimulating neurite length and number, and soma size to a comparable extent as BDNF, and that the increase in total neurite length is ascribed to axon outgrowth.
Collapse
|
9
|
Shpakov AO, Derkach KV, Zharova OA, Shpakova EA. The functional activity of the adenylate cyclase system in the brains of rats with metabolic syndrome induced by immunization with peptide 11–25 of the type 4 melanocortin receptor. NEUROCHEM J+ 2015. [DOI: 10.1134/s1819712415010092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
10
|
Irwin M, Greig A, Tvrdik P, Lucero MT. PACAP modulation of calcium ion activity in developing granule cells of the neonatal mouse olfactory bulb. J Neurophysiol 2014; 113:1234-48. [PMID: 25475351 DOI: 10.1152/jn.00594.2014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Ca(2+) activity in the CNS is critical for the establishment of developing neuronal circuitry prior to and during early sensory input. In developing olfactory bulb (OB), the neuromodulators that enhance network activity are largely unknown. Here we provide evidence that pituitary adenylate cyclase-activating peptide (PACAP)-specific PAC1 receptors (PAC1Rs) expressed in postnatal day (P)2-P5 mouse OB are functional and enhance network activity as measured by increases in calcium in genetically identified granule cells (GCs). We used confocal Ca(2+) imaging of OB slices from Dlx2-tdTomato mice to visualize GABAergic GCs. To address whether the PACAP-induced Ca(2+) oscillations were direct or indirect effects of PAC1R activation, we used antagonists for the GABA receptors (GABARs) and/or glutamate receptors (GluRs) in the presence and absence of PACAP. Combined block of GABARs and GluRs yielded a 66% decrease in the numbers of PACAP-responsive cells, suggesting that 34% of OB neurons are directly activated by PACAP. Similarly, immunocytochemistry using anti-PAC1 antibody showed that 34% of OB neurons express PAC1R. Blocking either GluRs or GABARs alone indirectly showed that PACAP stimulates release of both glutamate and GABA, which activate GCs. The appearance of PACAP-induced Ca(2+) activity in immature GCs suggests a role for PACAP in GC maturation. To conclude, we find that PACAP has both direct and indirect effects on neonatal OB GABAergic cells and may enhance network activity by promoting glutamate and GABA release. Furthermore, the numbers of PACAP-responsive GCs significantly increased between P2 and P5, suggesting that PACAP-induced Ca(2+) activity contributes to neonatal OB development.
Collapse
Affiliation(s)
- Mavis Irwin
- Department of Physiology, University of Utah School of Medicine, Salt Lake City, Utah
| | - Ann Greig
- Department of Physiology, University of Utah School of Medicine, Salt Lake City, Utah
| | - Petr Tvrdik
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah; Interdepartmental Neuroscience Program, University of Utah School of Medicine, Salt Lake City, Utah; and
| | - Mary T Lucero
- Department of Physiology, University of Utah School of Medicine, Salt Lake City, Utah; Interdepartmental Neuroscience Program, University of Utah School of Medicine, Salt Lake City, Utah; and Department of Neuroscience and Physiology, American University of the Caribbean, Cupecoy, Sint Maarten, Netherlands Antilles
| |
Collapse
|
11
|
An enriched environment ameliorates memory impairments in PACAP-deficient mice. Behav Brain Res 2014; 272:269-78. [DOI: 10.1016/j.bbr.2014.07.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 06/30/2014] [Accepted: 07/02/2014] [Indexed: 12/21/2022]
|
12
|
Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) Signalling Enhances Osteogenesis in UMR-106 Cell Line. J Mol Neurosci 2014; 54:555-73. [DOI: 10.1007/s12031-014-0389-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 07/22/2014] [Indexed: 01/14/2023]
|
13
|
Ago Y, Tanaka T, Ota Y, Kitamoto M, Imoto E, Takuma K, Matsuda T. Social crowding in the night-time reduces an anxiety-like behavior and increases social interaction in adolescent mice. Behav Brain Res 2014; 270:37-46. [DOI: 10.1016/j.bbr.2014.04.047] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/25/2014] [Accepted: 04/28/2014] [Indexed: 11/30/2022]
|
14
|
Huang H, Wang H, Figueiredo-Pereira ME. Regulating the ubiquitin/proteasome pathway via cAMP-signaling: neuroprotective potential. Cell Biochem Biophys 2014; 67:55-66. [PMID: 23686612 DOI: 10.1007/s12013-013-9628-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The cAMP-signaling pathway has been under intensive investigation for decades. It is a wonder that such a small simple molecule like cAMP can modulate a vast number of diverse processes in different types of cells. The ubiquitous involvement of cAMP-signaling in a variety of cellular events requires tight spatial and temporal control of its generation, propagation, compartmentalization, and elimination. Among the various steps of the cAMP-signaling pathway, G-protein-coupled receptors, adenylate cyclases, phosphodiesterases, the two major cAMP targets, i.e., protein kinase A and exchange protein activated by cAMP, as well as the A-kinase anchoring proteins, are potential targets for drug development. Herein we review the recent progress on the regulation and manipulation of different steps of the cAMP-signaling pathway. We end by focusing on the emerging role of cAMP-signaling in modulating protein degradation via the ubiquitin/proteasome pathway. New discoveries on the regulation of the ubiquitin/proteasome pathway by cAMP-signaling support the development of new therapeutic approaches to prevent proteotoxicity in chronic neurodegenerative disorders and other human disease conditions associated with impaired protein turnover by the ubiquitin/proteasome pathway and the accumulation of ubiquitin-protein aggregates.
Collapse
Affiliation(s)
- He Huang
- Department of Biological Sciences, Hunter College and Graduate Center, City University of New York, 695 Park Avenue, New York, NY 10065, USA
| | | | | |
Collapse
|
15
|
Chen BH, Yan BC, Park JH, Ahn JH, Lee DH, Kim IH, Cho JH, Lee JC, Kim SK, Lee B, Cho JH, Won MH, Lee YL. Aripiprazole, an atypical antipsychotic drug, improves maturation and complexity of neuroblast dendrites in the mouse dentate gyrus via increasing superoxide dismutases. Neurochem Res 2013; 38:1980-8. [PMID: 23836293 DOI: 10.1007/s11064-013-1104-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Revised: 06/27/2013] [Accepted: 06/28/2013] [Indexed: 01/02/2023]
Abstract
Apripiprazole (APZ) is well known as an atypical antipsychotic and antidepressant. In the present study, we investigated effects of APZ on cell proliferation and neuronal differentiation in the dentate gyrus (DG) of the adolescent mouse using BruU, Ki-67 and doublecortin (DCX) immunohistochemistry. BruU, Ki-67 and DCX-positive (+) cells were easily detected in the subgranular zone of the DG in the vehicle- and APZ-treated group. We found that in the 8 mg/kg APZ-treated group numbers of Ki-67(+), DCX(+) and BrdU(+)/DCX(+) cells were significantly increased compared with those in the vehicle-treated group. We also found that maturation and complexity of DCX(+) dendrites in the 8 mg/kg APZ-treated group was well improved compared with those in the vehicle-treated group. In addition, markedly decreased lipid peroxidation and increased superoxide dismutase 2 (SOD2) level were observed in the DG of the 8 mg/kg APZ-treated group. Our present findings indicate that APZ can enhance cell proliferation and neuroblast differentiation, particularly maturation and complexity of neuroblast dendrites, in the DG via decreasing lipid peroxidation and increasing SOD2 level.
Collapse
Affiliation(s)
- Bai Hui Chen
- Department of Physiology, Institute of Neurodegeneration and Neuroregeneration, College of Medicine, Hallym University, Chuncheon, 200-702, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
The importance of adult neurogenesis has only recently been accepted, resulting in a completely new field of investigation within stem cell biology. The regulation and functional significance of adult neurogenesis is currently an area of highly active research. G-protein-coupled receptors (GPCRs) have emerged as potential modulators of adult neurogenesis. GPCRs represent a class of proteins with significant clinical importance, because approximately 30% of all modern therapeutic treatments target these receptors. GPCRs bind to a large class of neurotransmitters and neuromodulators such as norepinephrine, dopamine, and serotonin. Besides their typical role in cellular communication, GPCRs are expressed on adult neural stem cells and their progenitors that relay specific signals to regulate the neurogenic process. This review summarizes the field of adult neurogenesis and its methods and specifies the roles of various GPCRs and their signal transduction pathways that are involved in the regulation of adult neural stem cells and their progenitors. Current evidence supporting adult neurogenesis as a model for self-repair in neuropathologic conditions, adult neural stem cell therapeutic strategies, and potential avenues for GPCR-based therapeutics are also discussed.
Collapse
Affiliation(s)
- Van A Doze
- Department of Molecular Cardiology, NB50, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | | |
Collapse
|
17
|
PACAP is an Endogenous Protective Factor—Insights from PACAP-Deficient Mice. J Mol Neurosci 2012; 48:482-92. [DOI: 10.1007/s12031-012-9762-0] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 03/22/2012] [Indexed: 01/07/2023]
|
18
|
Castorina A, Giunta S, Scuderi S, D'Agata V. Involvement of PACAP/ADNP signaling in the resistance to cell death in malignant peripheral nerve sheath tumor (MPNST) cells. J Mol Neurosci 2012; 48:674-83. [PMID: 22454142 DOI: 10.1007/s12031-012-9755-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 03/16/2012] [Indexed: 11/30/2022]
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are sarcomas able to grow under conditions of metabolic stress caused by insufficient nutrients or oxygen. Both pituitary adenylate cyclase-activating polypeptide (PACAP) and activity-dependent neuroprotective protein (ADNP) have glioprotective potential. However, whether PACAP/ADNP signaling is involved in the resistance to cell death in MPNST cells remains to be clarified. Here, we investigated the involvement of this signaling system in the survival response of MPNST cells against hydrogen peroxide (H(2)O(2))-evoked death both in the presence of normal serum (NS) and in serum-starved (SS) cells. Results showed that ADNP levels increased time-dependently (6-48 h) in SS cells. Treatment with PACAP38 (10(-9) to 10(-5) M) dose-dependently increased ADNP levels in NS but not in SS cells. PAC(1)/VPAC receptor antagonists completely suppressed PACAP-stimulated ADNP increase and partially reduced ADNP expression in SS cells. NS-cultured cells exposed to H(2)O(2) showed significantly reduced cell viability (~50 %), increased p53 and caspase-3, and DNA fragmentation, without affecting ADNP expression. Serum starvation significantly reduced H(2)O(2)-induced detrimental effects in MPNST cells, which were not further ameliorated by PACAP38. Altogether, these finding provide evidence for the involvement of an endogenous PACAP-mediated ADNP signaling system that increases MPNST cell resistance to H(2)O(2)-induced death upon serum starvation.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Apoptosis/physiology
- Culture Media, Serum-Free/pharmacology
- DNA Replication
- DNA, Neoplasm/analysis
- Dose-Response Relationship, Drug
- Gene Expression Regulation, Neoplastic/drug effects
- Hydrogen Peroxide/toxicity
- Neoplasm Proteins/physiology
- Nerve Sheath Neoplasms/pathology
- Nerve Tissue Proteins/biosynthesis
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/physiology
- Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology
- Pituitary Adenylate Cyclase-Activating Polypeptide/physiology
- Rats
- Real-Time Polymerase Chain Reaction
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I/antagonists & inhibitors
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I/physiology
- Receptors, Vasoactive Intestinal Peptide, Type II/antagonists & inhibitors
- Receptors, Vasoactive Intestinal Peptide, Type II/physiology
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Tumor Cells, Cultured/drug effects
- Tumor Cells, Cultured/metabolism
- Tumor Cells, Cultured/pathology
Collapse
Affiliation(s)
- Alessandro Castorina
- Department of Bio-Medical Sciences, University of Catania, Via S.Sofia 87, 95123, Catania, Italy
| | | | | | | |
Collapse
|
19
|
Yan BC, Yoo KY, Park JH, Lee CH, Choi JH, Won MH. The high dosage of earthworm (Eisenia andrei) extract decreases cell proliferation and neuroblast differentiation in the mouse hippocampal dentate gyrus. Anat Cell Biol 2011; 44:218-25. [PMID: 22025974 PMCID: PMC3195826 DOI: 10.5115/acb.2011.44.3.218] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 07/23/2011] [Accepted: 07/29/2011] [Indexed: 11/30/2022] Open
Abstract
Earthworm extract has shown anticancer characteristics. In the present study, we examined the effect of chronic treatment with a high dose of earthworm (Eisenia andrei) extract (EE) on cell proliferation and neuroblast differentiation in the hippocampal dentate gyrus (DG) of 3-week-old mice using 5-bromo-2'-deoxyuridine (BrdU) and Ki-67 immunohistochemistry for cell proliferation and doublecortin (DCX) immunohistochemistry for neuroblast differentiation, respectively. BrdU-, Ki-67-, and DCX-immunoreactive cells were easily detected in the subgranular zone of the DG in vehicle (saline)-treated mice. However, BrdU-, Ki-67-, and DCX-immunoreactive cells in the 500 mg/kg EE-treated mice decreased distinctively compared to those in the vehicle-treated mice. In addition, brain-derived neurotrophic factor (BDNF) immunoreactivity and its protein level decreased markedly in the DG of the EE-treated group compared to those in the vehicle-treated group. These results indicate that chronic treatment with high dose EE decreased cell proliferation and neuroblast differentiation, and that BDNF immunoreactivity decreased in the DG of EE-treated mice.
Collapse
Affiliation(s)
- Bing Chun Yan
- Department of Neurobiology, Kangwon National University School of Medicine, Chuncheon, Korea
| | | | | | | | | | | |
Collapse
|
20
|
Takuma K, Ago Y, Matsuda T. Preventive effects of an enriched environment on rodent psychiatric disorder models. J Pharmacol Sci 2011; 117:71-6. [PMID: 21881295 DOI: 10.1254/jphs.11r07cp] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
Interplay between genetic and environmental factors plays a key role in psychiatric disorders, as well as other brain diseases, cancer, and metabolic syndrome. In accordance with epidemiological findings, animal studies have pointed out the importance of a variety of environmental factors, such as viral infection during pregnancy or infancy, early parental loss or separation, and physical or sexual abuse in early life, in the etiology of psychiatric disorders. Conversely, positive effects of environmental factors against the pathogenesis of psychiatric disorders are also demonstrated, in which most of the animals are exposed to an "enriched environment". This review summarizes recent progress of research in this field focusing on the preventive effects of an "enriched environment" against the expression of behavioral abnormalities in rodent models of psychiatric disorders.
Collapse
Affiliation(s)
- Kazuhiro Takuma
- Laboratory of Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Japan
| | | | | |
Collapse
|