1
|
Mabondzo A, van de Kamp J, Mercimek-Andrews S. Dodecyl creatine ester therapy: from promise to reality. Cell Mol Life Sci 2024; 81:186. [PMID: 38632116 PMCID: PMC11024018 DOI: 10.1007/s00018-024-05218-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/26/2024] [Accepted: 03/05/2024] [Indexed: 04/19/2024]
Abstract
Pathogenic variants in SLC6A8, the gene which encodes creatine transporter SLC6A8, prevent creatine uptake in the brain and result in a variable degree of intellectual disability, behavioral disorders (e.g., autism spectrum disorder), epilepsy, and severe speech and language delay. There are no treatments to improve neurodevelopmental outcomes for creatine transporter deficiency (CTD). In this spotlight, we summarize recent advances in innovative molecules to treat CTD, with a focus on dodecyl creatine ester, the most promising drug candidate.
Collapse
Affiliation(s)
- Aloïse Mabondzo
- Paris Saclay University, CEA, Medicines and Healthcare Technologies Department (MTS), SPI, Neurovascular Unit Research and Therapeutic Innovation Laboratory, 91191, Gif-sur-Yvette cedex, France.
| | - Jiddeke van de Kamp
- Department of Human Genetics, Amsterdam UMC, Vrije Universtiteit Amsterdam, Amsterdam, The Netherlands
| | - Saadet Mercimek-Andrews
- Department of Medical Genetics, Faculty of Medicine and Dentistry, Neurosciences and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
2
|
Choi IY, Cha JH, Kim SY, Hsieh J, Cho KO. Seizure-induced LIN28A disrupts pattern separation via aberrant hippocampal neurogenesis. JCI Insight 2024; 9:e175627. [PMID: 38193536 PMCID: PMC10906464 DOI: 10.1172/jci.insight.175627] [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: 09/21/2023] [Accepted: 11/15/2023] [Indexed: 01/10/2024] Open
Abstract
Prolonged seizures can disrupt stem cell behavior in the adult hippocampus, an important brain structure for spatial memory. Here, using a mouse model of pilocarpine-induced status epilepticus (SE), we characterized spatiotemporal expression of Lin28a mRNA and proteins after SE. Unlike Lin28a transcripts, induction of LIN28A protein after SE was detected mainly in the subgranular zone, where immunoreactivity was found in progenitors, neuroblasts, and immature and mature granule neurons. To investigate roles of LIN28A in epilepsy, we generated Nestin-Cre:Lin28aloxP/loxP (conditional KO [cKO]) and Nestin-Cre:Lin28a+/+ (WT) mice to block LIN28A upregulation in all neuronal lineages after acute seizure. Adult-generated neuron- and hippocampus-associated cognitive impairments were absent in epileptic LIN28A-cKO mice, as evaluated by pattern separation and contextual fear conditioning tests, respectively, while sham-manipulated WT and cKO animals showed comparable memory function. Moreover, numbers of hilar PROX1-expressing ectopic granule cells (EGCs), together with PROX1+/NEUN+ mature EGCs, were significantly reduced in epileptic cKO mice. Transcriptomics analysis and IHC validation at 3 days after pilocarpine administration provided potential LIN28A downstream targets such as serotonin receptor 4. Collectively, our findings indicate that LIN28A is a potentially novel target for regulation of newborn neuron-associated memory dysfunction in epilepsy by modulating seizure-induced aberrant neurogenesis.
Collapse
Affiliation(s)
| | | | - Seong Yun Kim
- Department of Pharmacology, College of Medicine
- Department of Biomedicine & Health Sciences, and
- Catholic Neuroscience Institute, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jenny Hsieh
- Department of Neuroscience, Developmental and Regenerative Biology, and
- Brain Health Consortium, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - Kyung-Ok Cho
- Department of Pharmacology, College of Medicine
- Department of Biomedicine & Health Sciences, and
- Catholic Neuroscience Institute, The Catholic University of Korea, Seoul, Republic of Korea
- Institute for Aging and Metabolic Diseases and
- CMC Institute for Basic Medical Science, the Catholic Medical Center of The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
3
|
Mabondzo A, Harati R, Broca-Brisson L, Guyot AC, Costa N, Cacciante F, Putignano E, Baroncelli L, Skelton MR, Saab C, Martini E, Benech H, Joudinaud T, Gaillard JC, Armengaud J, Hamoudi R. Dodecyl creatine ester improves cognitive function and identifies key protein drivers including KIF1A and PLCB1 in a mouse model of creatine transporter deficiency. Front Mol Neurosci 2023; 16:1118707. [PMID: 37063368 PMCID: PMC10103630 DOI: 10.3389/fnmol.2023.1118707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/20/2023] [Indexed: 04/03/2023] Open
Abstract
Creatine transporter deficiency (CTD), a leading cause of intellectual disability is a result of the mutation in the gene encoding the creatine transporter SLC6A8, which prevents creatine uptake into the brain, causing mental retardation, expressive speech and language delay, autistic-like behavior and epilepsy. Preclinical in vitro and in vivo data indicate that dodecyl creatine ester (DCE) which increases the creatine brain content, might be a therapeutic option for CTD patients. To gain a better understanding of the pathophysiology and DCE treatment efficacy in CTD, this study focuses on the identification of biomarkers related to cognitive improvement in a Slc6a8 knockout mouse model (Slc6a8−/y) engineered to mimic the clinical features of CTD patients which have low brain creatine content. Shotgun proteomics analysis of 4,035 proteins in four different brain regions; the cerebellum, cortex, hippocampus (associated with cognitive functions) and brain stem, and muscle as a control, was performed in 24 mice. Comparison of the protein abundance in the four brain regions between DCE-treated intranasally Slc6a8−/y mice and wild type and DCE-treated Slc6a8−/y and vehicle group identified 14 biomarkers, shedding light on the mechanism of action of DCE. Integrative bioinformatics and statistical modeling identified key proteins in CTD, including KIF1A and PLCB1. The abundance of these proteins in the four brain regions was significantly correlated with both the object recognition and the Y-maze tests. Our findings suggest a major role for PLCB1, KIF1A, and associated molecules in the pathogenesis of CTD.
Collapse
Affiliation(s)
- Aloïse Mabondzo
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (MTS), Gif sur Yvette, France
- *Correspondence: Aloïse Mabondzo,
| | - Rania Harati
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharja, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Léa Broca-Brisson
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (MTS), Gif sur Yvette, France
| | - Anne-Cécile Guyot
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (MTS), Gif sur Yvette, France
| | - Narciso Costa
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (MTS), Gif sur Yvette, France
| | | | - Elena Putignano
- Institute of Neuroscience, National Research Council (CNR), Pisa, Italy
| | - Laura Baroncelli
- Institute of Neuroscience, National Research Council (CNR), Pisa, Italy
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, Pisa, Italy
| | - Matthew R. Skelton
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children’s Research Foundation, Cincinnati, OH, United States
| | - Cathy Saab
- Université de Paris and Université Paris Saclay, CEA, Stabilité Génétique Cellules Souches et Radiations, Fontenay aux Roses, France
| | - Emmanuelle Martini
- Université de Paris and Université Paris Saclay, CEA, Stabilité Génétique Cellules Souches et Radiations, Fontenay aux Roses, France
| | | | | | - Jean-Charles Gaillard
- Université Paris Saclay, CEA, Département Médicaments et Technologies pour la Santé (MTS), INRAE, Bagnol sur Cèze, France
| | - Jean Armengaud
- Université Paris Saclay, CEA, Département Médicaments et Technologies pour la Santé (MTS), INRAE, Bagnol sur Cèze, France
| | - Rifat Hamoudi
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
| |
Collapse
|
4
|
Song B, Kim CH. Cell-autonomous PLCβ1 modulation of neural stem/progenitor cell proliferation during adult hippocampal neurogenesis. Neurosci Lett 2022; 791:136899. [DOI: 10.1016/j.neulet.2022.136899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/14/2022] [Accepted: 09/30/2022] [Indexed: 10/31/2022]
|
5
|
Hamoudi W, Tripathi MK, Ojha SK, Amal H. A cross-talk between nitric oxide and the glutamatergic system in a Shank3 mouse model of autism. Free Radic Biol Med 2022; 188:83-91. [PMID: 35716826 DOI: 10.1016/j.freeradbiomed.2022.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 11/21/2022]
Abstract
Nitric oxide (NO) is a multifunctional signaling molecule that plays a crucial role in synaptic transmission and neuronal function. Pioneering studies show that nitric oxide (NO) and S-nitrosylation (SNO, the NO-mediated posttranslational modification) can engender nitrosative stress in the brain, contributing to neurodegenerative diseases. Little is known, however, about the aberrant NO signaling in neurodevelopmental disorders including autism spectrum disorder (ASD). We have recently shown that the Shank3 mutation in mice representing a model of ASD causes excessive NO levels and aberrant protein SNO. The glutamatergic system is involved in ASD, specifically in SHANK3 pathology. We used SNOTRAP technology to identify the SNO-proteome in the brain of the Shank3 mutant mice to understand the role of SNO in the glutamatergic system during the development of these mice. We conducted a systems biology analysis of the SNO-proteome to investigate the biological processes and signaling pathways in the brain of juvenile and adult Shank3 mutant and wild-type mice. The Shank3 mutation caused significant SNO-enrichment of a glutamate signaling pathway in the juvenile and adult mutant mice, although different protein subsets were S-nitrosylated in both ages. Cellular compartments analysis showed that "glutamatergic Synapse" is SNO-enriched significantly in the mutant mice of both ages. We also found eight S-nitrosylated proteins involved in glutamate transmission in both ages. 38 SNO-proteins found in the mutant mice are among the high-risk SFARI gene list. Biochemical examination shows a reduction in the levels of NMDA Receptor (NR1) in the cortex and striatum of the mutant mice of both ages. Neuronal NOS knockdown in SHSY-5Y rescued NR1 levels. In conclusion, this study reveals novel SNO of key glutamatergic proteins in Shank3 mutant mice and a cross-talk between nitric oxide and the glutamatergic system.
Collapse
Affiliation(s)
- Wajeha Hamoudi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Manish Kumar Tripathi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Shashank Kumar Ojha
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel.
| |
Collapse
|
6
|
Osawa K, Nakanishi Y, Noguchi M, Sugeno A, Goshima Y, Ohshima T. CRMP4 is required for the positioning and maturation of newly generated neurons in adult mouse hippocampus. Neurosci Lett 2022; 773:136503. [PMID: 35122931 DOI: 10.1016/j.neulet.2022.136503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/26/2021] [Accepted: 01/30/2022] [Indexed: 11/28/2022]
Abstract
Adult neurogenesis is a phenomenon in which neural stem cells differentiate and mature to generate new neurons in the adult brain. In mammals, the sites where adult neurogenesis occurs are limited to the subgranular zone (SGZ) of the hippocampal dentate gyrus and the subventricular zone. In the hippocampus, newly generated neurons migrate into the granule cell layer (GCL) and are integrated into neural circuits. Previous studies have revealed that CRMP4, a member of the CRMP family, is expressed in immature neurons in the hippocampal SGZ of the adult brain. However, the role of CRMP4 in adult neurogenesis is unknown. To study the role of CRMP4 in hippocampal adult neurogenesis, we compared adult neurogenesis between wild type and CRMP4-/- mice. In CRMP4-/- mice, the number of doublecortin (DCX)-positive cells was comparable to that in wild-type mice, and some DCX-positive cells were ectopically located in the granule cell layer, suggesting that CRMP4 is involved in the migration of adult neurogenesis. In addition, the number of calretinin-positive new neurons in the SGZ was significantly increased, whereas the number of EdU/NeuN-double positive neurons was decreased in CRMP4-/- mice, suggesting that CRMP4 plays an important role in neuronal maturation. Because CRMP4 is expressed in immature neurons, its expression may regulate the migration from the SGZ to the GCL during neuronal maturation in hippocampal adult neurogenesis.
Collapse
Affiliation(s)
- Koki Osawa
- Department of Life Science and Medical Bio-Science, Waseda University, Shinjuku-ku, Tokyo, 162-8480 Japan
| | - Yurika Nakanishi
- Department of Life Science and Medical Bio-Science, Waseda University, Shinjuku-ku, Tokyo, 162-8480 Japan
| | - Masahito Noguchi
- Department of Life Science and Medical Bio-Science, Waseda University, Shinjuku-ku, Tokyo, 162-8480 Japan
| | - Ayaka Sugeno
- Department of Life Science and Medical Bio-Science, Waseda University, Shinjuku-ku, Tokyo, 162-8480 Japan
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004 Japan
| | - Toshio Ohshima
- Department of Life Science and Medical Bio-Science, Waseda University, Shinjuku-ku, Tokyo, 162-8480 Japan.
| |
Collapse
|
7
|
Doi H, Matsuda T, Sakai A, Matsubara S, Hoka S, Yamaura K, Nakashima K. Early-life midazolam exposure persistently changes chromatin accessibility to impair adult hippocampal neurogenesis and cognition. Proc Natl Acad Sci U S A 2021; 118:e2107596118. [PMID: 34526402 PMCID: PMC8463898 DOI: 10.1073/pnas.2107596118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2021] [Indexed: 01/02/2023] Open
Abstract
Linkage between early-life exposure to anesthesia and subsequent learning disabilities is of great concern to children and their families. Here we show that early-life exposure to midazolam (MDZ), a widely used drug in pediatric anesthesia, persistently alters chromatin accessibility and the expression of quiescence-associated genes in neural stem cells (NSCs) in the mouse hippocampus. The alterations led to a sustained restriction of NSC proliferation toward adulthood, resulting in a reduction of neurogenesis that was associated with the impairment of hippocampal-dependent memory functions. Moreover, we found that voluntary exercise restored hippocampal neurogenesis, normalized the MDZ-perturbed transcriptome, and ameliorated cognitive ability in MDZ-exposed mice. Our findings thus explain how pediatric anesthesia provokes long-term adverse effects on brain function and provide a possible therapeutic strategy for countering them.
Collapse
Affiliation(s)
- Hiroyoshi Doi
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
| | - Taito Matsuda
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan;
| | - Atsuhiko Sakai
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
| | - Shuzo Matsubara
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
| | - Sumio Hoka
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
- Department of Pharmaceutical Sciences, International University of Health and Welfare, 831-8501 Fukuoka, Japan
| | - Ken Yamaura
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
| | - Kinichi Nakashima
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan;
| |
Collapse
|
8
|
Kunii Y, Matsumoto J, Izumi R, Nagaoka A, Hino M, Shishido R, Sainouchi M, Akatsu H, Hashizume Y, Kakita A, Yabe H. Evidence for Altered Phosphoinositide Signaling-Associated Molecules in the Postmortem Prefrontal Cortex of Patients with Schizophrenia. Int J Mol Sci 2021; 22:8280. [PMID: 34361045 PMCID: PMC8348881 DOI: 10.3390/ijms22158280] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/24/2021] [Accepted: 07/28/2021] [Indexed: 12/26/2022] Open
Abstract
Phosphoinositides (PIs) play important roles in the structure and function of the brain. Associations between PIs and the pathophysiology of schizophrenia have been studied. However, the significance of the PI metabolic pathway in the pathology of schizophrenia is unknown. We examined the expression of PI signaling-associated proteins in the postmortem brain of schizophrenia patients. Protein expression levels of phosphatidylinositol 4-phosphate 5-kinase type-1 gamma (PIP5K1C), phosphatidylinositol 4-kinase alpha (PIK4CA, also known as PIK4A), phosphatase and tensin homolog deleted from chromosome 10 (PTEN), protein kinase B (Akt), and glycogen synthase kinase 3β (GSK3β) were measured using enzyme-linked immunosorbent assays and multiplex fluorescent bead-based immunoassays of the prefrontal cortex (PFC) of postmortem samples from 23 schizophrenia patients and 47 normal controls. We also examined the association between PIK4CA expression and its genetic variants in the same brain samples. PIK4CA expression was lower, whereas Akt expression was higher, in the PFC of schizophrenia patients than in that of controls; PIP5K1C, PTEN, and GSK3β expression was not different. No single-nucleotide polymorphism significantly affected protein expression. We identified molecules involved in the pathology of schizophrenia via this lipid metabolic pathway. These results suggest that PIK4CA is involved in the mechanism underlying the pathogenesis of schizophrenia and is a potential novel therapeutic target.
Collapse
Affiliation(s)
- Yasuto Kunii
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan; (J.M.); (R.I.); (A.N.); (M.H.); (R.S.); (H.Y.)
- Department of Disaster Psychiatry, International Research Institute of Disaster Science, Tohoku University, Sendai 980-8573, Japan
| | - Junya Matsumoto
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan; (J.M.); (R.I.); (A.N.); (M.H.); (R.S.); (H.Y.)
| | - Ryuta Izumi
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan; (J.M.); (R.I.); (A.N.); (M.H.); (R.S.); (H.Y.)
| | - Atsuko Nagaoka
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan; (J.M.); (R.I.); (A.N.); (M.H.); (R.S.); (H.Y.)
| | - Mizuki Hino
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan; (J.M.); (R.I.); (A.N.); (M.H.); (R.S.); (H.Y.)
- Department of Disaster Psychiatry, International Research Institute of Disaster Science, Tohoku University, Sendai 980-8573, Japan
| | - Risa Shishido
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan; (J.M.); (R.I.); (A.N.); (M.H.); (R.S.); (H.Y.)
| | - Makoto Sainouchi
- Department of Pathology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (M.S.); (A.K.)
| | - Hiroyasu Akatsu
- Department of Community-Based Medical Education, Nagoya City University Graduate School of Medical Science, Aichi 467-8601, Japan;
- Department of Community-Based Medicine, Nagoya City University Graduate School of Medical Science, Aichi 467-8601, Japan
- Choju Medical Institute, Fukushimura Hospital, Aichi 441-8124, Japan;
| | - Yoshio Hashizume
- Choju Medical Institute, Fukushimura Hospital, Aichi 441-8124, Japan;
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (M.S.); (A.K.)
| | - Hirooki Yabe
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan; (J.M.); (R.I.); (A.N.); (M.H.); (R.S.); (H.Y.)
| |
Collapse
|
9
|
Zhao Z, Li B, Wu Y, Chen X, Guo Y, Shen Y, Huang H. Ketamine affects the integration of developmentally generated granule neurons in the adult stage. BMC Neurosci 2019; 20:60. [PMID: 31852437 PMCID: PMC6921590 DOI: 10.1186/s12868-019-0542-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 12/07/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Ketamine has been reported to cause neonatal neurotoxicity in a variety of developing animal models. Various studies have been conducted to study the mechanism of neurotoxicity for general anesthetic use during the neonatal period. Previous experiments have suggested that developmentally generated granule neurons in the hippocampus dentate gyrus (DG) supported hippocampus-dependent memory. Therefore, this study aimed to investigate whether ketamine affects the functional integration of developmentally generated granule neurons in the DG. For this purpose,the postnatal day 7 (PND-7) Sprague-Dawley (SD) rats were divided into the control group and the ketamine group (rats who received 4 injections of 40 mg/kg ketamine at 1 h intervals). To label dividing cells, BrdU was administered for three consecutive days after the ketamine exposure; NeuN+/BrdU+cells were observed by using immunofluorescence. To evaluate the developmentally generated granule neurons that support hippocampus-dependent memory, spatial reference memory was tested by using Morris Water Maze at 3 months old, after which the immunofluorescence was used to detect c-Fos expression in the NeuN+/BrdU+ cells. The expression of caspase-3 was measured by western blot to detect the apoptosis in the hippocampal DG. RESULTS The present results showed that the neonatal ketamine exposure did not influence the survival rate of developmentally generated granule neurons at 2 and 3 months old, but ketamine interfered with the integration of these neurons into the hippocampal DG neural circuits and caused a deficit in hippocampal-dependent spatial reference memory tasks. CONCLUSIONS In summary, these findings may promote more studies to investigate the neurotoxicity of ketamine in the developing brain.
Collapse
Affiliation(s)
- Zhanqiang Zhao
- Department of Anesthesiology, Jiangning Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Bing Li
- Department of Anesthesiology, Jiangning Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Yuqing Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China
| | - Xujun Chen
- Department of Anesthesiology, Jiangning Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Yan Guo
- Department of Anesthesiology, Jiangning Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Yang Shen
- Department of Anesthesiology, Jiangning Hospital of Traditional Chinese Medicine, Nanjing, China
| | - He Huang
- Department of Anesthesiology, First Affiliated Hospital With Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu, People's Republic of China.
| |
Collapse
|
10
|
Morgan JA, Singhal G, Corrigan F, Jaehne EJ, Jawahar MC, Baune BT. TNF signalling via the TNF receptors mediates the effects of exercise on cognition-like behaviours. Behav Brain Res 2018; 353:74-82. [PMID: 29969604 DOI: 10.1016/j.bbr.2018.06.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 06/29/2018] [Accepted: 06/29/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Altered TNF levels are associated with cognitive impairment in depression, schizophrenia, bipolar disorder, and Alzheimer's disease (AD). Exercise improves cognition-like behaviours, reduces the expression of tumour necrosis factor alpha (TNF), and increases expression of the soluble TNF receptors soluble TNFR1 (sTNFR1) and sTNFR2. We suggest TNF and its receptors are involved in cognitive function and dysfunction, and investigate whether exercise mediates its effects on cognitive function via TNF and its receptors. METHODS We utilised C57BL/6, TNF-/-, TNFR1-/-, and TNFR2-/- mice to compare exercise to non-exercise control groups to investigate whether exercise exerts its effects on various types of cognition-like behaviours via TNF and its receptors. RESULTS Recognition memory improved with exercise in WT mice, was impaired in TNFR1-/- exercise mice, showed non-significant impairment with exercise in TNF-/- mice, and no changes in TNFR2-/- mice. In spatial learning there were exercise related improvements in WT mice, non-significant but meaningful impairments evident in TNFR1-/- exercise mice, modest improvement in TNF-/- exercise mice, and potentially meaningful non-significant improvements in TNFR2-/- exercise mice. Moreover, WT and TNFR2-/- mice displayed noteworthy non-significant improvements in spatial memory, whereas TNFR1-/- exercise mice demonstrated non-significant spatial memory impairment. Exercise did not alter cognitive flexibility in any strain. DISCUSSION TNF receptor signalling via the TNFR1 and TNFR2 appears to mediate the effects of exercise on cognitive-like behaviours. The potential for exercise to regulate human TNF and TNF signalling and cognitive dysfunction needs investigation under inflammatory conditions including depression and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Julie A Morgan
- The University of Adelaide, School of Medicine, Discipline of Psychiatry, Adelaide, Australia
| | - Gaurav Singhal
- The University of Adelaide, School of Medicine, Discipline of Psychiatry, Adelaide, Australia
| | - Frances Corrigan
- The University of Adelaide, School of Medicine, Discipline of Medical Sciences, Adelaide, Australia
| | - Emily J Jaehne
- The University of Adelaide, School of Medicine, Discipline of Psychiatry, Adelaide, Australia; La Trobe University, School of Psychology and Public Health, Melbourne, Australia
| | - Magdalene C Jawahar
- The University of Adelaide, School of Medicine, Discipline of Psychiatry, Adelaide, Australia
| | - Bernhard T Baune
- The University of Adelaide, School of Medicine, Discipline of Psychiatry, Adelaide, Australia.
| |
Collapse
|
11
|
Garwain O, Valla K, Scarlata S. Phospholipase Cβ1 regulates proliferation of neuronal cells. FASEB J 2018; 32:2891-2898. [PMID: 29401590 DOI: 10.1096/fj.201701284r] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cells have developed lineage-specific mechanisms to control proliferation and drive morphologic changes upon differentiation. A hallmark of differentiation is the assembly of signaling molecules that transduce extracellular signals, such as the production of the G protein-regulated enzyme phospholipase Cβ (PLCβ), which generates calcium signals from sensory stimuli. We found that in most cancerous cell lines there is positive correlation between PLCβ1 levels and cell proliferation. In cells of neuronal lineage, however, reducing PLCβ1 levels increases the rate of proliferation. Using a combination of biochemical and biophysical methods, we find that, in the G1 phase, a cytosolic population of PLCβ1 associates with cyclin-dependent kinase 16 (CDK16), a neuron-specific enzyme that is activated by cyclin Y to inactivate the antioncogenic protein p27Kip1. Binding of PLCβ1 directly inhibits CDK16 activity and in turn reduces the ability of cells to enter the S phase. Activation of Gαq by carbachol causes movement of PLCβ from the cytosol to the plasma membrane, reducing its association with CDK16. Similarly, the overexpression of activated Gαq moves PLCβ1 to the membrane, reverses G1 arrest, and promotes proliferation, thereby connecting external stimuli with cell proliferation. Our results present a model in which the transient high expression of PLCβ1 that occurs at the onset of differentiation arrests cells in the G1 phase through its association with CDK16 and allows CDK16 to transition to its postmitotic function of neurite outgrowth and trafficking of synaptic vesicles. The novel role of PLCβ1 in neuronal cell proliferation offers a unique interaction that can be manipulated to guide cells into a neuronal phenotype or to develop therapies for neuroblastomas.-Garwain, O., Valla, K., Scarlata, S. Phospholipase Cβ1 regulates proliferation of neuronal cells.
Collapse
Affiliation(s)
- Osama Garwain
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Kaitlyn Valla
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Suzanne Scarlata
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts, USA.,Department of Physiology and Biophysics, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
12
|
Han J, Kim HJ, Schafer ST, Paquola A, Clemenson GD, Toda T, Oh J, Pankonin AR, Lee BS, Johnston ST, Sarkar A, Denli AM, Gage FH. Functional Implications of miR-19 in the Migration of Newborn Neurons in the Adult Brain. Neuron 2017; 91:79-89. [PMID: 27387650 DOI: 10.1016/j.neuron.2016.05.034] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 04/13/2016] [Accepted: 05/18/2016] [Indexed: 12/27/2022]
Abstract
Altered microRNA profiles have been implicated in human brain disorders. However, the functional contribution of individual microRNAs to neuronal development and function is largely unknown. Here, we report biological functions for miR-19 in adult neurogenesis. We determined that miR-19 is enriched in neural progenitor cells (NPCs) and downregulated during neuronal development in the adult hippocampus. By manipulating miR-19 in NPCs for gain- and loss-of-function studies, we discovered that miR-19 regulates cell migration by directly targeting Rapgef2. Concordantly, dysregulation of miR-19 in NPCs alters the positioning of newborn neurons in the adult brain. Furthermore, we found abnormal expression of miR-19 in human NPCs generated from schizophrenic patient-derived induced pluripotent stem cells (iPSCs) that have been described as displaying aberrant migration. Our study demonstrates the significance of posttranscriptional gene regulation by miR-19 in preventing the irregular migration of adult-born neurons that may contribute to the etiology of schizophrenia.
Collapse
Affiliation(s)
- Jinju Han
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Hyung Joon Kim
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Simon T Schafer
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; Institute of Physiology, University of Greifswald, 17495 Karlsburg, Germany
| | - Apua Paquola
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Gregory D Clemenson
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Tomohisa Toda
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jinseo Oh
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Aimee R Pankonin
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Bo Suk Lee
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Stephen T Johnston
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Anindita Sarkar
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Ahmet M Denli
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Fred H Gage
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
13
|
Acevedo-Triana CA, Rojas MJ, Cardenas FP. Running wheel training does not change neurogenesis levels or alter working memory tasks in adult rats. PeerJ 2017; 5:e2976. [PMID: 28503368 PMCID: PMC5426350 DOI: 10.7717/peerj.2976] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 01/10/2017] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Exercise can change cellular structure and connectivity (neurogenesis or synaptogenesis), causing alterations in both behavior and working memory. The aim of this study was to evaluate the effect of exercise on working memory and hippocampal neurogenesis in adult male Wistar rats using a T-maze test. METHODS An experimental design with two groups was developed: the experimental group (n = 12) was subject to a forced exercise program for five days, whereas the control group (n = 9) stayed in the home cage. Six to eight weeks after training, the rats' working memory was evaluated in a T-maze test and four choice days were analyzed, taking into account alternation as a working memory indicator. Hippocampal neurogenesis was evaluated by means of immunohistochemistry of BrdU positive cells. RESULTS No differences between groups were found in the behavioral variables (alternation, preference index, time of response, time of trial or feeding), or in the levels of BrdU positive cells. DISCUSSION Results suggest that although exercise may have effects on brain structure, a construct such as working memory may require more complex changes in networks or connections to demonstrate a change at behavioral level.
Collapse
Affiliation(s)
| | - Manuel J. Rojas
- Animal Health Department, Universidad Nacional de Colombia, Bogotá, Colombia
| | | |
Collapse
|
14
|
Udawela M, Scarr E, Boer S, Um JY, Hannan AJ, McOmish C, Felder CC, Thomas EA, Dean B. Isoform specific differences in phospholipase C beta 1 expression in the prefrontal cortex in schizophrenia and suicide. NPJ SCHIZOPHRENIA 2017; 3:19. [PMID: 28560265 PMCID: PMC5441535 DOI: 10.1038/s41537-017-0020-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 03/09/2017] [Accepted: 03/21/2017] [Indexed: 11/09/2022]
Abstract
Our previous study demonstrated that phospholipase C beta 1 mRNA was down-regulated in Brodmann's area 46 from subjects with schizophrenia. However, phospholipase C beta 1 protein has also been shown to be lower in Brodmann's area 8 and 9 from teenage suicide subjects, creating a potential confound in interpreting the findings in schizophrenia due to the high suicide rate associated with this disorder. To begin to reconcile and consolidate these findings, in this study, we measured mRNA and protein levels of phospholipase C beta 1 variants a and b in Brodmann's area 46 and Brodmann's area 9 from subjects with schizophrenia, many of whom were suicide completers, and determined the diagnostic specificity of observed findings. Consistent with our previous study, levels of phospholipase C beta 1 a and b mRNA, but not protein, were lower in Brodmann's area 46 from subjects with schizophrenia. In Brodmann's area 9, phospholipase C beta 1a protein levels were lower in subjects with schizophrenia, while phospholipase C beta 1b mRNA was higher and protein was lower in those that had died of suicide. Altered protein levels in Brodmann's area 9 appeared to be diagnostically specific, as we did not detect these changes in subjects with bipolar disorder, major depressive disorder or suicide completers with no diagnosis of mental illness. We further assessed the relationship between phospholipase C beta 1 and levels of muscarinic receptors (CHRMs) that signal through this protein, in both human and Chrm knockout mouse central nervous system tissue, and found no strong relationship between the two. Understanding central nervous system differences in downstream effector pathways in schizophrenia may lead to improved treatment strategies and help to identify those at risk of suicide.
Collapse
Affiliation(s)
- M Udawela
- Molecular Psychiatry Laboratories, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC Australia
| | - E Scarr
- Molecular Psychiatry Laboratories, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC Australia
- Department of Psychiatry, University of Melbourne, Parkville, VIC Australia
| | - S Boer
- Molecular Psychiatry Laboratories, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC Australia
| | - J Y Um
- Molecular Psychiatry Laboratories, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC Australia
- Cardiovascular and Neurology Products Division, Drug Evaluation Department, National Institute of Food and Drug Safety Evaluation, Osong Health Technology Administration Complex, Chungcheongbuk-do, South Korea
| | - A J Hannan
- Epigenetics and Neural Plasticity Laboratory, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC Australia
| | - C McOmish
- Molecular Psychiatry Laboratories, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC Australia
| | - C C Felder
- Lilly Research Laboratories, Neuroscience Research Division, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN USA
| | - E A Thomas
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA USA
| | - B Dean
- Molecular Psychiatry Laboratories, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC Australia
- Cardiovascular and Neurology Products Division, Drug Evaluation Department, National Institute of Food and Drug Safety Evaluation, Osong Health Technology Administration Complex, Chungcheongbuk-do, South Korea
| |
Collapse
|
15
|
Matsumoto J, Nakanishi H, Kunii Y, Sugiura Y, Yuki D, Wada A, Hino M, Niwa SI, Kondo T, Waki M, Hayasaka T, Masaki N, Akatsu H, Hashizume Y, Yamamoto S, Sato S, Sasaki T, Setou M, Yabe H. Decreased 16:0/20:4-phosphatidylinositol level in the post-mortem prefrontal cortex of elderly patients with schizophrenia. Sci Rep 2017; 7:45050. [PMID: 28332626 PMCID: PMC5362900 DOI: 10.1038/srep45050] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 02/17/2017] [Indexed: 01/17/2023] Open
Abstract
The etiology of schizophrenia includes phospholipid abnormalities. Phospholipids are bioactive substances essential for brain function. To analyze differences in the quantity and types of phospholipids present in the brain tissue of patients with schizophrenia, we performed a global analysis of phospholipids in multiple brain samples using liquid chromatography electrospray ionization mass/mass spectrometry (LC-ESI/MS/MS) and imaging mass spectrometry (IMS). We found significantly decreased 16:0/20:4-phosphatidylinositol (PI) levels in the prefrontal cortex (PFC) in the brains from patients with schizophrenia in the LC-ESI/MS/MS, and that the 16:0/20:4-PI in grey matter was most prominently diminished according to the IMS experiments. Previous reports investigating PI pathology of schizophrenia did not identify differences in the sn-1 and sn-2 fatty acyl chains. This study is the first to clear the fatty acid composition of PI in brains from patients with schizophrenia. Alteration in the characteristic fatty acid composition of PI may also affect neuronal function, and could play a role in the etiology of schizophrenia. Although further studies are necessary to understand the role of reduced 16:0/20:4-PI levels within the prefrontal cortex in the etiology of schizophrenia, our results provide insight into the development of a novel therapy for the clinical treatment of schizophrenia.
Collapse
Affiliation(s)
- Junya Matsumoto
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, Fukushima 960-1295, Japan
| | - Hiroki Nakanishi
- Research Center for Biosignal, Akita University, 1-1-1 Hondo, Akita, Akita 010-8543, Japan
- Akita Lipid Technologies, LLC.,1-2, Nukazuka, Yanagida, Akita, 010-0825, Japan
| | - Yasuto Kunii
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, Fukushima 960-1295, Japan
- Department of Psychiatry, Aizu Medical Center, Fukushima Medical University, 21-2 Maeda, Yazawa Kawahigashimachi, Aizuwakamatsu, Fukushima 969-3492, Japan
| | - Yuki Sugiura
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Dai Yuki
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Akira Wada
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, Fukushima 960-1295, Japan
- Department of Neuropsychiatry, The University of Tokyo Hospital, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Mizuki Hino
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, Fukushima 960-1295, Japan
| | - Shin-Ichi Niwa
- Department of Psychiatry, Aizu Medical Center, Fukushima Medical University, 21-2 Maeda, Yazawa Kawahigashimachi, Aizuwakamatsu, Fukushima 969-3492, Japan
| | - Takeshi Kondo
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Michihiko Waki
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Takahiro Hayasaka
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Noritaka Masaki
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Hiroyasu Akatsu
- Choju Medical Institute, Fukushimura Hospital, 19-14 Yamanaka, Noyori-cho, Toyohashi, Aichi 441-8124, Japan
- Department of Community-based Medical Education/Department of Community-based Medicine, Nagoya City University Graduate School of Medical Science, 1, Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Yoshio Hashizume
- Choju Medical Institute, Fukushimura Hospital, 19-14 Yamanaka, Noyori-cho, Toyohashi, Aichi 441-8124, Japan
| | - Sakon Yamamoto
- Choju Medical Institute, Fukushimura Hospital, 19-14 Yamanaka, Noyori-cho, Toyohashi, Aichi 441-8124, Japan
| | - Shinji Sato
- Choju Medical Institute, Fukushimura Hospital, 19-14 Yamanaka, Noyori-cho, Toyohashi, Aichi 441-8124, Japan
- Quests Research Institute, Otsuka Pharmaceutical Co. Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima, Tokushima 771-0192, Japan
| | - Takehiko Sasaki
- Research Center for Biosignal, Akita University, 1-1-1 Hondo, Akita, Akita 010-8543, Japan
- Akita Lipid Technologies, LLC.,1-2, Nukazuka, Yanagida, Akita, 010-0825, Japan
- Department of Medical Biology Graduate School of Medicine, Akita University, 1-1-1 Hondo, Akita, Tokushima 010-8543, Japan
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- Department of Anatomy, The university of Hong Kong, 6/F, William MW Mong Block 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
- Division of Neural Systematics, National Institute for Physiological Sciences, 38 Nishigonaka Myodaiji, Okazaki, Aichi, 444-8585, Japan
- Riken Center for Molecular Imaging Science, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Hirooki Yabe
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, Fukushima 960-1295, Japan
| |
Collapse
|
16
|
Han J, Gage FH. A role for miR-19 in the migration of adult-born neurons and schizophrenia. NEUROGENESIS 2016; 3:e1251873. [PMID: 28405585 PMCID: PMC5384614 DOI: 10.1080/23262133.2016.1251873] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 10/13/2016] [Accepted: 10/18/2016] [Indexed: 12/14/2022]
Abstract
The latest miRNA database (Release 21) annotated 2588 and 1915 miRNAs in the human and mouse genomes, respectively.1 However, the biological roles of miRNAs in vivo remain largely unknown. In particular, the physiological and pathological roles of individual microRNAs in the brain have not been investigated extensively although expression profiles of microRNAs have been reported in many given conditions. In a recent study,2 we identified miR-19, which is enriched in adult hippocampal neural progenitor cells (NPCs), as a key regulator for adult hippocampal neurogenesis. miR-19 is an intrinsic factor regulating the migration of newborn neurons by modulating expression level of RAPGEF2. After observing the abnormal expression patterns of miR-19 and RAPGEF2 in NPCs derived from induced pluripotent stem cells of schizophrenic patients, which display aberrant cell migration, we proposed miR-19 as a molecule associated with schizophrenia. The results illustrate that a single microRNA has the potential to impact the functions of the brain. Identifying miRNA-mediated posttranscriptional gene regulation in the brain will expand our understanding of brain development and functions and the etiologies of several brain disorders.
Collapse
Affiliation(s)
- Jinju Han
- The Salk Institute for Biological Sciences , La Jolla, CA, USA
| | - Fred H Gage
- The Salk Institute for Biological Sciences , La Jolla, CA, USA
| |
Collapse
|
17
|
Nuclear Lipids in the Nervous System: What they do in Health and Disease. Neurochem Res 2016; 42:321-336. [PMID: 27766461 DOI: 10.1007/s11064-016-2085-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/21/2016] [Accepted: 10/11/2016] [Indexed: 12/18/2022]
Abstract
In the last 20 years it has been widely demonstrated that cell nucleus contains neutral and polar lipids localized in nuclear membranes, nucleoli, nuclear matrix and chromatin. Nuclear lipids may show specific organization forming nuclear lipid microdomains and have both structural and functional roles. Depending on their localization, nuclear lipids play different roles such as the regulation of nuclear membrane and nuclear matrix fluidity but they also can act as platforms for vitamin and hormone function, for active chromatin anchoring, and for the regulation of gene expression, DNA duplication and transcription. Crosstalk among different kinds of lipid signalling pathways influence the physiopathology of numerous cell types. In neural cells the nuclear lipids are involved in cell proliferation, differentiation, inflammation, migration and apoptosis. Abnormal metabolism of nuclear lipids might be closely associated with tumorigenesis and neurodegenerative diseases such as Alzheimer disease and Parkinson disease among others.
Collapse
|
18
|
Ng T, Hor CHH, Chew B, Zhao J, Zhong Z, Ryu JR, Goh ELK. Neuropilin 2 Signaling Is Involved in Cell Positioning of Adult-born Neurons through Glycogen Synthase Kinase-3β (GSK3β). J Biol Chem 2016; 291:25088-25095. [PMID: 27687730 DOI: 10.1074/jbc.m116.755215] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 09/27/2016] [Indexed: 11/06/2022] Open
Abstract
Proper positioning of neurons is fundamental for brain functions. However, little is known on how adult-born neurons generated in the hilar side of hippocampal dentate gyrus migrate into the granular cell layer. Because class 3 Semaphorins (Sema3) are involved in dendritic growth of these newborn neurons, we examined whether they are essential for cell positioning. We disrupted Sema3 signaling by silencing neuropilin 1 (NRP1) or 2 (NRP2), the main receptors for Sema3A and Sema3F, in neural progenitors of adult mouse dentate gyrus. Silencing of NRP2, but not NRP1, affected cell positioning of adult newborn neurons. Glycogen synthase kinase-3β (GSK3β) knockdown phenocopied this NRP2 silencing-mediated cell positioning defect, but did not affect dendritic growth. Furthermore, GSK3β is activated upon stimulation with Sema3F, and GSK3β overexpression rescued the cell positioning phenotypes seen in NRP2-deficient neurons. These results point to a new role for NRP2 in the positioning of neurons during adult hippocampal neurogenesis, acting via the GSK3β signaling pathway.
Collapse
Affiliation(s)
- Teclise Ng
- From the Programme in Neuroscience and Behavioral Disorder and
| | - Catherine H H Hor
- Neuroscience Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore.,the Department of Research, National Neuroscience Institute, Singapore 308433, Singapore
| | - Benjamin Chew
- From the Programme in Neuroscience and Behavioral Disorder and
| | - Jing Zhao
- GlaxoSmithKline (China) R&D Co., Ltd., Shanghai 201203, China
| | - Zhong Zhong
- GlaxoSmithKline (China) R&D Co., Ltd., Shanghai 201203, China
| | - Jae Ryun Ryu
- From the Programme in Neuroscience and Behavioral Disorder and
| | - Eyleen L K Goh
- From the Programme in Neuroscience and Behavioral Disorder and .,Neuroscience Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore.,the Department of Research, National Neuroscience Institute, Singapore 308433, Singapore.,the Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore, and.,the KK Research Center, KK Women's and Children's Hospital, Singapore 229899, Singapore
| |
Collapse
|
19
|
GABAergic Regulation of Adult Hippocampal Neurogenesis. Mol Neurobiol 2016; 54:5497-5510. [DOI: 10.1007/s12035-016-0072-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/18/2016] [Indexed: 01/17/2023]
|
20
|
Huang H, Liu CM, Sun J, Hao T, Xu CM, Wang D, Wu YQ. Ketamine Affects the Neurogenesis of the Hippocampal Dentate Gyrus in 7-Day-Old Rats. Neurotox Res 2016; 30:185-98. [PMID: 26966008 DOI: 10.1007/s12640-016-9615-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/22/2016] [Accepted: 03/01/2016] [Indexed: 01/20/2023]
Abstract
Ketamine has been reported to cause neonatal neurotoxicity via a neuronal apoptosis mechanism; however, no in vivo research has reported whether ketamine could affect postnatal neurogenesis in the hippocampal dentate gyrus (DG). A growing number of experiments suggest that postnatal hippocampal neurogenesis is the foundation of maintaining normal hippocampus function into adulthood. Therefore, this study investigated the effect of ketamine on hippocampal neurogenesis. Male Sprague-Dawley rats were divided into two groups: the control group (equal volume of normal saline), and the ketamine-anesthesia group (40 mg/kg ketamine in four injections at 1 h intervals). The S-phase marker 5-bromodeoxyuridine (BrdU) was administered after ketamine exposure to postnatal day 7 (PND-7) rats, and the neurogenesis in the hippocampal DG was assessed using single- or double-immunofluorescence staining. The expression of GFAP in the hippocampal DG was measured by western blot analysis. Spatial reference memory was tested by Morris water maze at 2 months after PND-7 rats exposed to ketamine treatment. The present results showed that neonatal ketamine exposure significantly inhibited neural stem cell (NSC) proliferation, decreased astrocytic differentiation, and markedly enhanced neuronal differentiation. The disruptive effect of ketamine on the proliferation and differentiation of NSCs lasted at least 1 week and disappeared by 2 weeks after ketamine exposure. Moreover, the migration of newborn neurons in the granule cell layer and the growth of astrocytes in the hippocampal DG were inhibited by ketamine on PND-37 and PND-44. Finally, ketamine caused a deficit in hippocampal-dependent spatial reference memory tasks at 2 months old. Our results suggested that ketamine may interfere with hippocampal neurogenesis and long-term neurocognitive function in PND-7 rats. These findings may provide a new perspective to explain the adult neurocognitive dysfunction induced by neonatal ketamine exposure.
Collapse
Affiliation(s)
- He Huang
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Cun-Ming Liu
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Sun
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ting Hao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou, China
| | - Chun-Mei Xu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou, China
| | - Dan Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou, China
| | - Yu-Qing Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou, China.
| |
Collapse
|
21
|
Shea CJA, Carhuatanta KAK, Wagner J, Bechmann N, Moore R, Herman JP, Jankord R. Variable impact of chronic stress on spatial learning and memory in BXD mice. Physiol Behav 2015; 150:69-77. [PMID: 26079812 DOI: 10.1016/j.physbeh.2015.06.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 06/04/2015] [Accepted: 06/11/2015] [Indexed: 12/15/2022]
Abstract
The effects of chronic stress on learning are highly variable across individuals. This variability stems from gene-environment interactions. However, the mechanisms by which stress affects genetic predictors of learning are unclear. Thus, we aim to determine whether the genetic pathways that predict spatial memory performance are altered by previous exposure to chronic stress. Sixty-two BXD recombinant inbred strains of mice, as well as parent strains C57BL/6J and DBA/2J, were randomly assigned as behavioral control or to a chronic variable stress paradigm and then underwent behavioral testing to assess spatial memory and learning performance using the Morris water maze. Quantitative trait loci (QTL) mapping was completed for average escape latency times for both control and stress animals. Loci on chromosomes 5 and 10 were found in both control and stress environmental populations; eight additional loci were found to be unique to either the control or stress environment. In sum, results indicate that certain genetic loci predict spatial memory performance regardless of prior stress exposure, while exposure to stress also reveals unique genetic predictors of training during the memory task. Thus, we find that genetic predictors contributing to spatial learning and memory are susceptible to the presence of chronic stress.
Collapse
Affiliation(s)
- Chloe J A Shea
- Applied Neuroscience, 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson AFB, OH 45433, United States
| | - Kimberly A K Carhuatanta
- Applied Neuroscience, 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson AFB, OH 45433, United States; Research Associate Program, National Research Council, National Academies of Science, Washington DC 20001, United States
| | - Jessica Wagner
- Applied Neuroscience, 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson AFB, OH 45433, United States
| | - Naomi Bechmann
- Applied Neuroscience, 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson AFB, OH 45433, United States; Infoscitex, Inc., Dayton, OH 45435, United States
| | - Raquel Moore
- Applied Neuroscience, 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson AFB, OH 45433, United States; Infoscitex, Inc., Dayton, OH 45435, United States
| | - James P Herman
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45267, United States
| | - Ryan Jankord
- Applied Neuroscience, 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson AFB, OH 45433, United States.
| |
Collapse
|
22
|
Keshavan MS, Mehta UM, Padmanabhan JL, Shah JL. Dysplasticity, metaplasticity, and schizophrenia: Implications for risk, illness, and novel interventions. Dev Psychopathol 2015; 27:615-35. [PMID: 25997775 PMCID: PMC6283269 DOI: 10.1017/s095457941500019x] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In this paper, we review the history of the concept of neuroplasticity as it relates to the understanding of neuropsychiatric disorders, using schizophrenia as a case in point. We briefly review the myriad meanings of the term neuroplasticity, and its neuroscientific basis. We then review the evidence for aberrant neuroplasticity and metaplasticity associated with schizophrenia as well as the risk for developing this illness, and discuss the implications of such understanding for prevention and therapeutic interventions. We argue that the failure and/or altered timing of plasticity of critical brain circuits might underlie cognitive and deficit symptoms, and may also lead to aberrant plastic reorganization in other circuits, leading to affective dysregulation and eventually psychosis. This "dysplastic" model of schizophrenia can suggest testable etiology and treatment-relevant questions for the future.
Collapse
Affiliation(s)
- Matcheri S. Keshavan
- Department of Psychiatry, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States
| | - Urvakhsh Meherwan Mehta
- Department of Psychiatry, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States
- Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Jaya L. Padmanabhan
- Department of Psychiatry, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States
| | - Jai L. Shah
- Douglas Hospital Research Center and Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
23
|
Aberrant hippocampal neurogenesis contributes to epilepsy and associated cognitive decline. Nat Commun 2015; 6:6606. [PMID: 25808087 PMCID: PMC4375780 DOI: 10.1038/ncomms7606] [Citation(s) in RCA: 320] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/10/2015] [Indexed: 12/18/2022] Open
Abstract
Acute seizures after a severe brain insult can often lead to epilepsy and cognitive impairment. Aberrant hippocampal neurogenesis follows the insult but the role of adult-generated neurons in the development of chronic seizures or associated cognitive deficits remains to be determined. Here we show that the ablation of adult neurogenesis before pilocarpine-induced acute seizures in mice leads to a reduction in chronic seizure frequency. We also show that ablation of neurogenesis normalizes epilepsy-associated cognitive deficits. Remarkably, the effect of ablating adult neurogenesis before acute seizures is long lasting as it suppresses chronic seizure frequency for nearly 1 year. These findings establish a key role of neurogenesis in chronic seizure development and associated memory impairment and suggest that targeting aberrant hippocampal neurogenesis may reduce recurrent seizures and restore cognitive function following a pro-epileptic brain insult. Aberrant hippocampal neurogenesis often occurs after acute seizures that produce epilepsy and cognitive impairment but the role of neurogenesis in the development of epilepsy is unclear. Here the authors suppress adult neurogenesis in mice preceding seizures and show that it reduces subsequent chronic seizure frequency and epilepsy-associated cognitive decline.
Collapse
|
24
|
Zhang Z, van Praag H. Maternal immune activation differentially impacts mature and adult-born hippocampal neurons in male mice. Brain Behav Immun 2015; 45:60-70. [PMID: 25449671 DOI: 10.1016/j.bbi.2014.10.010] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 10/14/2014] [Accepted: 10/16/2014] [Indexed: 12/30/2022] Open
Abstract
Schizophrenia is associated with deficits in the hippocampus, a brain area important for learning and memory. The dentate gyrus (DG) of the hippocampus develops both before and after birth. To study the relative contribution of mature and adult-born DG granule cells to disease etiology, we compared both cell populations in a mouse model of psychiatric illness resulting from maternal immune activation. Polyriboinosinic-polyribocytidilic acid (PolyIC, 5mg/kg) or saline was given on gestation day 15 to pregnant female C57Bl/6 mice. Male offspring (n=105), was administered systemic bromodeoxyuridine (BrdU, 50mg/kg) (n=52) or intracerebral retroviral injection into the DG (n=53), to label dividing cells at one month of age. Two months later behavioral tests were performed to evaluate disease phenotype. Immunohistochemistry and whole-cell patch clamping were used to assess morphological and physiological characteristics of DG cells. Three-month-old PolyIC exposed male offspring exhibited deficient pre-pulse inhibition, spatial maze performance and motor coordination, as well as increased depression-like behavior. Histological analysis showed reduced DG volume and parvalbumin positive interneuron number. Both mature and new hippocampal neurons showed modifications in intrinsic properties such as increased input resistance and lower current threshold, and decreased action potential number. Reduced GABAergic inhibitory transmission was observed only in mature DG neurons. Differential impairments in mature DG cells and adult-born new neurons may have implications for behavioral deficits associated with maternal immune activation.
Collapse
Affiliation(s)
- Zhi Zhang
- Neuroplasticity and Behavior Unit, Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Henriette van Praag
- Neuroplasticity and Behavior Unit, Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
25
|
Ransome MI. Could androgens maintain specific domains of mental health in aging men by preserving hippocampal neurogenesis? Neural Regen Res 2014; 7:2227-39. [PMID: 25538744 PMCID: PMC4268723 DOI: 10.3969/j.issn.1673-5374.2012.028.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 07/10/2012] [Indexed: 12/18/2022] Open
Abstract
Interest surrounds the role of sex-hormones in regulating brain function outside of reproductive behaviour. Declining androgen production in aging males has been associated with cognitive impairment, depression and increased risk of developing Alzheimer's disease. Indication for testosterone replacement therapy is based on biochemically determined low circulating testosterone combined with manifest symptoms. However, which aspects of age-related cognitive decline are attributable to low circulating testosterone remain ambiguous. Studies examining cognition in aging men receiving testosterone replacement therapy have yielded equivocal results. The exact role of testosterone in maintaining cognitive function and the underlying neural mechanisms are largely unknown, though it would appear to be domain specific. Clarity in this area will provide clinical direction toward addressing an increasing healthcare burden of mental health decline coincident with increasing longevity. The premise that androgens contribute to maintaining aspects of mental health in aging men by preserving hippocampal neurogenesis will be used as a forum in this review to discuss current knowledge and the need for further studies to better define testosterone replacement strategies for aging male health.
Collapse
Affiliation(s)
- Mark I Ransome
- Florey Neurosciences Institute, Melbourne Brain Centre, the University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
26
|
Vakalopoulos C. The effect of deficient muscarinic signaling on commonly reported biochemical effects in schizophrenia and convergence with genetic susceptibility loci in explaining symptom dimensions of psychosis. Front Pharmacol 2014; 5:277. [PMID: 25566074 PMCID: PMC4266038 DOI: 10.3389/fphar.2014.00277] [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: 08/19/2014] [Accepted: 11/27/2014] [Indexed: 11/13/2022] Open
Abstract
With the advent of DSM 5 criticism has generally centered on a lack of biological validity of the diagnostic criteria. Part of the problem in describing a nosology of psychosis is the tacit assumption of multiple genetic causes each with an incremental loading on the clinical picture that fails to differentiate a clear underlying pathophysiology of high impact. The aim of this paper is to consolidate a primary theory of deficient muscarinic signaling underlying key clinical features of schizophrenia and its regulation by several important genetic associations including neuregulin, DISC and dysbindin. Secondary reductions in markers for GABAergic function and changes in the levels of interneuron calcium binding proteins parvalbumin and calbindin can be attributed to dysfunctional muscarinic transduction. A parallel association exists for cytokine production. The convergent pathway hypothesis is likewise used to model dopaminergic and glutamatergic theories of schizophrenia. The negative symptom dimension is correlated with dysfunction of Akt and ERK transduction, a major point of convergence. The present paradigm predicts the importance of a recent finding of a deletion in a copy number variant of PLCB1 and its potential use if replicated, as one of the first testable biological markers differentiating schizophrenia from bipolar disorder and further subtyping of schizophrenia into deficit and non-deficit. Potential limitations of PLCB1 as a prospective marker are also discussed.
Collapse
|
27
|
Philip F, Sahu S, Caso G, Scarlata S. Role of phospholipase C-β in RNA interference. Adv Biol Regul 2013; 53:319-330. [PMID: 23916604 PMCID: PMC3805765 DOI: 10.1016/j.jbior.2013.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Revised: 07/03/2013] [Accepted: 07/04/2013] [Indexed: 06/02/2023]
Abstract
Phospholipase C-β (PLCβ) enzymes are activated by G proteins in response to agents such as hormones and neurotransmitters, and have been implicated in leukemias and neurological disorders. PLCβ activity causes an increase in intracellular calcium which ultimately leads to profound changes in the cell. PLCβ localizes to three cellular compartments: the plasma membrane, the cytosol and the nucleus. Under most cell conditions, the majority of PLCβ localizes to the plasma membrane where it interacts with G proteins. In trying to determine the factors that localize PLCβ to the cytosol and nucleus, we have recently identified the binding partner, TRAX. TRAX is a nuclease and part of the machinery involved in RNA interference. This review discusses the interaction between PLCβ and TRAX, and its repercussions in G protein signaling and RNA silencing.
Collapse
Affiliation(s)
- Finly Philip
- Department of Physiology & Biophysics, Stony Brook University, Stony Brook, NY 11794-8661, USA
| | | | | | | |
Collapse
|
28
|
Knockdown of the glucocorticoid receptor alters functional integration of newborn neurons in the adult hippocampus and impairs fear-motivated behavior. Mol Psychiatry 2013; 18:993-1005. [PMID: 22925833 DOI: 10.1038/mp.2012.123] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2011] [Revised: 06/28/2012] [Accepted: 07/05/2012] [Indexed: 01/24/2023]
Abstract
Glucocorticoids (GCs) secreted after stress reduce adult hippocampal neurogenesis, a process that has been implicated in cognitive aspects of psychopathology, amongst others. Yet, the exact role of the GC receptor (GR), a key mediator of GC action, in regulating adult neurogenesis is largely unknown. Here, we show that GR knockdown, selectively in newborn cells of the hippocampal neurogenic niche, accelerates their neuronal differentiation and migration. Strikingly, GR knockdown induced ectopic positioning of a subset of the new granule cells, altered their dendritic complexity and increased their number of mature dendritic spines and mossy fiber boutons. Consistent with the increase in synaptic contacts, cells with GR knockdown exhibit increased basal excitability parallel to impaired contextual freezing during fear conditioning. Together, our data demonstrate a key role for the GR in newborn hippocampal cells in mediating their synaptic connectivity and structural as well as functional integration into mature hippocampal circuits involved in fear memory consolidation.
Collapse
|
29
|
Sabbagh JJ, Murtishaw AS, Bolton MM, Heaney CF, Langhardt M, Kinney JW. Chronic ketamine produces altered distribution of parvalbumin-positive cells in the hippocampus of adult rats. Neurosci Lett 2013; 550:69-74. [PMID: 23827228 DOI: 10.1016/j.neulet.2013.06.040] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 05/23/2013] [Accepted: 06/20/2013] [Indexed: 02/03/2023]
Abstract
The underlying mechanisms of schizophrenia pathogenesis are not well understood. Increasing evidence supports the glutamatergic hypothesis that posits a hypofunction of the N-methyl D-aspartate (NMDA) receptor on specific gamma amino-butyric acid (GABA)-ergic neurons may be responsible for the disorder. Alterations in the GABAergic system have been observed in schizophrenia, most notably a change in the expression of parvalbumin (PV) in the cortex and hippocampus. Several reports also suggest abnormal neuronal migration may play a role in the etiology of schizophrenia. The current study examined the positioning and distribution of PV-positive cells in the hippocampus following chronic treatment with the NMDA receptor antagonist ketamine. A robust increase was found in the number of PV-positive interneurons located outside the stratum oriens (SO), the layer where most of these cells are normally localized, as well as an overall numerical increase in CA3 PV cells. These results suggest ketamine leads to an abnormal distribution of PV-positive cells, which may be indicative of aberrant migratory activity and possibly related to the Morris water maze deficits observed. These findings may also be relevant to alterations observed in schizophrenia populations.
Collapse
Affiliation(s)
- Jonathan J Sabbagh
- Department of Psychology, University of Nevada Las Vegas, Las Vegas NV 89154, United States
| | | | | | | | | | | |
Collapse
|
30
|
A sensitive period for GABAergic interneurons in the dentate gyrus in modulating sensorimotor gating. J Neurosci 2013; 33:6691-704. [PMID: 23575865 DOI: 10.1523/jneurosci.0032-12.2013] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Developmental perturbations during adolescence have been hypothesized to be a risk factor for the onset of several neuropsychiatric diseases. However the physiological alterations that result from such insults are incompletely understood. We investigated whether a defined perturbation during adolescence affected hippocampus-dependent sensorimotor gating functions, a proposed endophenotype in several psychiatric diseases, most notably schizophrenia. The developmental perturbation was induced during adolescence in mice using an antimitotic agent, methylazoxymethanol acetate (MAM), during postnatal weeks (PW) 4-6. MAM-treated mice showed a decrease in hippocampal neurogenesis immediately after treatment, which was restored by PW10 in adulthood. However, the mice treated with MAM during adolescent stages exhibited a persistent sensorimotor gating deficiency and a reduction in prepulse inhibition-related activation of hippocampal and prefrontal neurons in adulthood. Cellular analyses found a reduction of GABAergic inhibitory neurons and abnormal dendritic morphology of immature neurons in the dentate gyrus (DG). Interestingly, bilateral infusion of muscimol, a GABAA receptor agonist, into the DG region reversed the prepulse inhibition abnormality in MAM-treated mice. Furthermore, the behavioral deficits together with the decrease in the number of GABAergic neurons in this MAM model were rescued by exposure to an enriched environment during a defined critical adolescent period. These observations suggest a possible role for GABAergic interneurons in the DG during adolescence. This role may be related to the establishment of neural circuitry required for sensorimotor gating. It is plausible that changes in neurogenesis during this window may affect the survival of GABAergic interneurons, although this link needs to be causally addressed.
Collapse
|
31
|
Sui Y, Vermeulen R, Hökfelt T, Horne MK, Stanić D. Female mice lacking cholecystokinin 1 receptors have compromised neurogenesis, and fewer dopaminergic cells in the olfactory bulb. Front Cell Neurosci 2013; 7:13. [PMID: 23459364 PMCID: PMC3584826 DOI: 10.3389/fncel.2013.00013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 02/03/2013] [Indexed: 12/17/2022] Open
Abstract
Neurogenesis in the adult rodent brain is largely restricted to the subependymal zone (SVZ) of the lateral ventricle and subgranular zone (SGZ) of the dentate gyrus (DG). We examined whether cholecystokinin (CCK) through actions mediated by CCK1 receptors (CCK1R) is involved in regulating neurogenesis. Proliferating cells in the SVZ, measured by 5-bromo-2-deoxyuridine (BrdU) injected 2 h prior to death or by immunoreactivity against Ki67, were reduced by 37 and 42%, respectively, in female (but not male) mice lacking CCK1Rs (CCK1R(-/-)) compared to wild-type (WT). Generation of neuroblasts in the SVZ and rostral migratory stream (RMS) was also affected, since the number of doublecortin (DCX)-immunoreactive (ir) neuroblasts in these regions decreased by 29%. In the SGZ of female CCK1R(-/-) mice, BrdU-positive (+), and Ki67-ir cells were reduced by 38 and 56%, respectively, while DCX-ir neuroblasts were down 80%. Subsequently, the effect of reduced SVZ/SGZ proliferation on the generation and survival of mature adult-born cells in female CCK1R(-/-) mice was examined. In the OB granule cell layer (GCL), the number of neuronal nuclei (NeuN)-ir and calretinin-ir cells was stable compared to WT, and 42 days after BrdU injections, the number of BrdU+ cells co-expressing GABA- or NeuN-like immunoreactivity (LI) was similar. Compared to WT, the granule cell layer of the DG in female CCK1R(-/-) mice had a similar number of calbindin-ir cells and BrdU+ cells co-expressing calbindin-LI 42 days after BrdU injections. However, the OB glomerular layer (GL) of CCK1R(-/-) female mice had 11% fewer NeuN-ir cells, 23% less TH-ir cells, and a 38% and 29% reduction in BrdU+ cells that co-expressed TH-LI or GABA-LI, respectively. We conclude that CCK, via CCK1Rs, is involved in regulating the generation of proliferating cells and neuroblasts in the adult female mouse brain, and mechanisms are in place to maintain steady neuronal populations in the OB and DG when the rate of proliferation is altered.
Collapse
Affiliation(s)
- Yi Sui
- Neurodegeneration Division, Florey Institute of Neuroscience and Mental Health, University of Melbourne Parkville, VIC, Australia ; Department of Neurology, Shenyang First People's Hospital Shenyang, China
| | | | | | | | | |
Collapse
|
32
|
Ransome MI, Hannan AJ. Impaired basal and running-induced hippocampal neurogenesis coincides with reduced Akt signaling in adult R6/1 HD mice. Mol Cell Neurosci 2013; 54:93-107. [PMID: 23384443 DOI: 10.1016/j.mcn.2013.01.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Revised: 01/24/2013] [Accepted: 01/25/2013] [Indexed: 10/27/2022] Open
Abstract
Huntington's disease (HD) is a fatal neurodegenerative disorder affecting a range of cellular and molecular functions in the brain. Deficits in adult hippocampal neurogenesis (AHN) have been documented in the R6/1 mouse model of HD. Here we examined basal and running-induced neuronal precursor proliferation in adult female and male R6/1 HD mice. We further tested whether sequential delivery of voluntary running followed by environmental enrichment could synergistically enhance functional AHN in female R6/1 HD mice. R6/1 HD mice engaged in significantly reduced levels of voluntary running, with males showing a more severe deficit. Basal neural precursor proliferation in the hippocampal sub-granular zone remained unchanged between female and male R6/1 HD mice and neither sex significantly responded to running-induced proliferation. While discrete provision of running wheels and enriched environments doubled AHN in adult female R6/1 HD mice it did not reflect the significant 3-fold increase in female wildtypes. Nevertheless, triple-label c-Fos/BrdU/NeuN immunofluorescence and confocal microscopy provided evidence that the doubling of AHN in female R6/1 HD mice was functional. Intrinsic cellular dysfunction mediated by protein aggregates containing mutant huntingtin (mHtt) did not appear to coincide with AHN deficits. In the hippocampus of female R6/1 HD mice, proliferating precursors and 6 week old adult-generated neurons were devoid of mHtt immuno-reactive aggregates, as were endothelial, microglial and astroglial cells populating the neurogenic niche. Serum transforming growth factor-β concentrations remained unaltered in female R6/1 HD mice as did the hippocampal levels of proliferating microglia and glial fibrillarly acidic protein expression. Examining the growth hormone/insulin-like growth factor 1 (GH/IGF-1) axis showed no change in base-line serum GH between genotypes. However, despite a reduced distance, acute running increases serum GH in both female wildtype and R6/1 HD mice. Serum IGF-1 levels were increased in female R6/1 HD mice compared to wildtypes during daytime inactive period, while hippocampal levels of the IGF-1 receptor remained unchanged. Running induced Akt phosphorylation in the hippocampus of female wildtype mice, which was not reflected in R6/1 HD mice. Total Akt levels were decreased in the hippocampus of both control and running R6/1 HD mice. Our results show adult-generated hippocampal neurons in female R6/1 HD mice express c-Fos and that running and Akt signaling deficits may mediate reduced basal and running-induced AHN levels.
Collapse
Affiliation(s)
- Mark I Ransome
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3010, Australia.
| | | |
Collapse
|
33
|
Fares RP, Belmeguenai A, Sanchez PE, Kouchi HY, Bodennec J, Morales A, Georges B, Bonnet C, Bouvard S, Sloviter RS, Bezin L. Standardized environmental enrichment supports enhanced brain plasticity in healthy rats and prevents cognitive impairment in epileptic rats. PLoS One 2013; 8:e53888. [PMID: 23342033 PMCID: PMC3544705 DOI: 10.1371/journal.pone.0053888] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 12/04/2012] [Indexed: 12/23/2022] Open
Abstract
Environmental enrichment of laboratory animals influences brain plasticity, stimulates neurogenesis, increases neurotrophic factor expression, and protects against the effects of brain insult. However, these positive effects are not constantly observed, probably because standardized procedures of environmental enrichment are lacking. Therefore, we engineered an enriched cage (the Marlau™ cage), which offers: (1) minimally stressful social interactions; (2) increased voluntary exercise; (3) multiple entertaining activities; (4) cognitive stimulation (maze exploration), and (5) novelty (maze configuration changed three times a week). The maze, which separates food pellet and water bottle compartments, guarantees cognitive stimulation for all animals. Compared to rats raised in groups in conventional cages, rats housed in Marlau™ cages exhibited increased cortical thickness, hippocampal neurogenesis and hippocampal levels of transcripts encoding various genes involved in tissue plasticity and remodeling. In addition, rats housed in Marlau™ cages exhibited better performances in learning and memory, decreased anxiety-associated behaviors, and better recovery of basal plasma corticosterone level after acute restraint stress. Marlau™ cages also insure inter-experiment reproducibility in spatial learning and brain gene expression assays. Finally, housing rats in Marlau™ cages after severe status epilepticus at weaning prevents the cognitive impairment observed in rats subjected to the same insult and then housed in conventional cages. By providing a standardized enriched environment for rodents during housing, the Marlau™ cage should facilitate the uniformity of environmental enrichment across laboratories.
Collapse
MESH Headings
- Adaptation, Psychological/physiology
- Animals
- Anxiety/complications
- Body Weight
- Brain/cytology
- Brain/pathology
- Brain/physiology
- Brain/physiopathology
- CA1 Region, Hippocampal/cytology
- CA1 Region, Hippocampal/pathology
- CA1 Region, Hippocampal/physiology
- CA1 Region, Hippocampal/physiopathology
- Cognition
- Eating
- Exploratory Behavior/physiology
- Health
- Housing, Animal/standards
- Lipid Metabolism
- Male
- Neurogenesis/genetics
- Neuronal Plasticity
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Reference Standards
- Reproducibility of Results
- Restraint, Physical/psychology
- Status Epilepticus/pathology
- Status Epilepticus/physiopathology
- Status Epilepticus/psychology
- Stress, Psychological/complications
- Stress, Psychological/pathology
- Stress, Psychological/physiopathology
- Synapses/pathology
Collapse
Affiliation(s)
- Raafat P. Fares
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Inserm, Institut National de la Santé et de la Recherche Médicale, U1028, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- CNRS, Centre National de la Recherche Scientifique, UMR5292, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- IDÉE, Institut Des ÉpilepsiEs, Lyon, France
- IRBA, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Amor Belmeguenai
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Inserm, Institut National de la Santé et de la Recherche Médicale, U1028, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- CNRS, Centre National de la Recherche Scientifique, UMR5292, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- IDÉE, Institut Des ÉpilepsiEs, Lyon, France
| | - Pascal E. Sanchez
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Inserm, Institut National de la Santé et de la Recherche Médicale, U1028, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- CNRS, Centre National de la Recherche Scientifique, UMR5292, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- IDÉE, Institut Des ÉpilepsiEs, Lyon, France
| | - Hayet Y. Kouchi
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Inserm, Institut National de la Santé et de la Recherche Médicale, U1028, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- CNRS, Centre National de la Recherche Scientifique, UMR5292, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- IDÉE, Institut Des ÉpilepsiEs, Lyon, France
| | - Jacques Bodennec
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Inserm, Institut National de la Santé et de la Recherche Médicale, U1028, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- CNRS, Centre National de la Recherche Scientifique, UMR5292, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- IDÉE, Institut Des ÉpilepsiEs, Lyon, France
| | - Anne Morales
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Inserm, Institut National de la Santé et de la Recherche Médicale, U1028, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- CNRS, Centre National de la Recherche Scientifique, UMR5292, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- IDÉE, Institut Des ÉpilepsiEs, Lyon, France
| | - Béatrice Georges
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Inserm, Institut National de la Santé et de la Recherche Médicale, U1028, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- CNRS, Centre National de la Recherche Scientifique, UMR5292, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- IDÉE, Institut Des ÉpilepsiEs, Lyon, France
| | - Chantal Bonnet
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Inserm, Institut National de la Santé et de la Recherche Médicale, U1028, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- CNRS, Centre National de la Recherche Scientifique, UMR5292, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- IDÉE, Institut Des ÉpilepsiEs, Lyon, France
| | - Sandrine Bouvard
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Inserm, Institut National de la Santé et de la Recherche Médicale, U1028, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- CNRS, Centre National de la Recherche Scientifique, UMR5292, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- IDÉE, Institut Des ÉpilepsiEs, Lyon, France
| | - Robert S. Sloviter
- Department of Neurobiology, Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Laurent Bezin
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Inserm, Institut National de la Santé et de la Recherche Médicale, U1028, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- CNRS, Centre National de la Recherche Scientifique, UMR5292, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- IDÉE, Institut Des ÉpilepsiEs, Lyon, France
- * E-mail:
| |
Collapse
|
34
|
Arnold SA, Hagg T. Serotonin 1A receptor agonist increases species- and region-selective adult CNS proliferation, but not through CNTF. Neuropharmacology 2012; 63:1238-47. [PMID: 22884499 PMCID: PMC3438376 DOI: 10.1016/j.neuropharm.2012.07.047] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 07/13/2012] [Accepted: 07/27/2012] [Indexed: 01/06/2023]
Abstract
Endogenous ciliary neurotrophic factor (CNTF)(1) regulates neurogenesis of the adult brain in the hippocampal subgranular zone (SGZ)(2) and the subventricular zone (SVZ)(3). We have previously shown that the cAMP-inhibiting D2 dopamine receptor increases neurogenesis by inducing astroglial CNTF expression. Here, we investigated the potential role of CNTF in the proliferative response to pharmacological stimulation of the serotonin 1A (5-HT1A)(4) receptor, which also inhibits cAMP, in adult mice and rats. Like others, we show that systemic treatment with the active R-enantiomer of the 5-HT1A agonist 8-Hydroxy-2-(di-n-propylamino)tetralin (8-OH-DPAT)(5) induces proliferation in the SGZ in rats using unbiased stereology of 5-Bromo-2'-deoxyuridine (BrdU)(6) positive nuclei. However, despite the bioactivity of R-8-OH-DPAT, as also shown by a decrease in hippocampal nNOS(7) mRNA levels, it did not increase CNTF mRNA as shown by highly specific quantitative RT-PCR (qPCR)(8). Surprisingly, R-8-OH-DPAT did not cause an increase in SVZ proliferation in rats or in either the SVZ or SGZ of two different strains of mice, C57BL/6J, and 129SvEv, using acute or chronic treatments. There also were no changes in CNTF mRNA, and also not in mice treated with a widely used racemic mixture of 8-OH-DPAT, higher doses or after intracerebral injection, which reduced nNOS. In contrast to the others, we propose that the 5-HT1A receptor might be non-functional in mice with regards to regulating normal neurogenesis and has region-selective activities in rats. These species- and region-specific actions raise important questions about the role of the 5-HT1A receptor in human neurogenesis and its implications for the field of depression.
Collapse
Affiliation(s)
- Sheila A. Arnold
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Kentucky 40292
- Departments of Neurological Surgery, University of Louisville, Kentucky 40292
| | - Theo Hagg
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Kentucky 40292
- Departments of Neurological Surgery, University of Louisville, Kentucky 40292
- Pharmacology and Toxicology, University of Louisville, Kentucky 40292
| |
Collapse
|
35
|
Schaeffer EL, Kühn F, Schmitt A, Gattaz WF, Gruber O, Schneider-Axmann T, Falkai P, Schmitt A. Increased cell proliferation in the rat anterior cingulate cortex following neonatal hypoxia: relevance to schizophrenia. J Neural Transm (Vienna) 2012; 120:187-95. [PMID: 22806004 PMCID: PMC3535379 DOI: 10.1007/s00702-012-0859-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Accepted: 06/29/2012] [Indexed: 01/20/2023]
Abstract
As a consequence of obstetric complications, neonatal hypoxia has been discussed as an environmental factor in the pathophysiology of schizophrenia. However, the biological consequences of hypoxia are unclear. The neurodevelopmental hypothesis of schizophrenia suggests that the onset of abnormal brain development and neuropathology occurs perinatally, whereas symptoms of the disease appear in early adulthood. In our animal model of chronic neonatal hypoxia, we have detected behavioral alterations resembling those known from schizophrenia. Disturbances in cell proliferation possibly contribute to the pathophysiology of this disease. In the present study, we used postnatal rats to investigate cell proliferation in several brain areas following neonatal hypoxia. Rats were repeatedly exposed to hypoxia (89 % N2, 11 % O2) from postnatal day (PD) 4–8. We then evaluated cell proliferation on PD 13 and 39, respectively. These investigations were performed in the anterior cingulate cortex (ACC), caudate-putamen (CPU), dentate gyrus, and subventricular zone. Rats exposed to hypoxia exhibited increased cell proliferation in the ACC at PD 13, normalizing at PD 39. In other brain regions, no alterations have been detected. Additionally, hypoxia-treated rats showed decreased CPU volume at PD 13. The results of the present study on the one hand support the assumption of chronic hypoxia influencing transient cell proliferation in the ACC, and on the other hand reveal normalization during ageing.
Collapse
Affiliation(s)
- Evelin L. Schaeffer
- Laboratory of Neuroscience (LIM-27), Department and Institute of Psychiatry, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Franziska Kühn
- Department of Psychiatry and Psychotherapy, Georg-August-University, von-Siebold-Str. 5, 37075 Göttingen, Germany
| | - Angelika Schmitt
- Department of Psychiatry and Psychotherapy, University of Würzburg, Füchsleinstr. 15, 97080 Würzburg, Germany
| | - Wagner F. Gattaz
- Laboratory of Neuroscience (LIM-27), Department and Institute of Psychiatry, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Oliver Gruber
- Department of Psychiatry and Psychotherapy, Georg-August-University, von-Siebold-Str. 5, 37075 Göttingen, Germany
| | - Thomas Schneider-Axmann
- Department of Psychiatry and Psychotherapy, Georg-August-University, von-Siebold-Str. 5, 37075 Göttingen, Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, Georg-August-University, von-Siebold-Str. 5, 37075 Göttingen, Germany
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University, Nußbaumstr. 7, 80336 Munich, Germany
| | - Andrea Schmitt
- Department of Psychiatry and Psychotherapy, Georg-August-University, von-Siebold-Str. 5, 37075 Göttingen, Germany
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University, Nußbaumstr. 7, 80336 Munich, Germany
| |
Collapse
|
36
|
Pang TYC, Hannan AJ. Enhancement of cognitive function in models of brain disease through environmental enrichment and physical activity. Neuropharmacology 2012; 64:515-28. [PMID: 22766390 DOI: 10.1016/j.neuropharm.2012.06.029] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 06/06/2012] [Accepted: 06/15/2012] [Indexed: 12/21/2022]
Abstract
This review will provide an overview of the non-drug based approaches that have been demonstrated to enhance cognitive function of the compromised brain, primarily focussed on the two most widely adopted paradigms of environmental enrichment and enhanced physical exercise. Environmental enrichment involves the generation of novelty and complexity in animal housing conditions which facilitates enhanced sensory and cognitive stimulation as well as physical activity. In a wide variety of animal models of brain disorders, environmental enrichment and exercise have been found to have beneficial effects, including cognitive enhancement, delayed disease onset, enhanced cellular plasticity and associated molecular processes. Potential cellular and molecular mechanisms will also be discussed, which have relevance for the future development of 'enviromimetics', drugs which could mimic or enhance the beneficial effects of environmental stimulation. This article is part of a Special Issue entitled 'Cognitive Enhancers'.
Collapse
Affiliation(s)
- Terence Y C Pang
- Florey Neuroscience Institutes, Melbourne Brain Centre, University of Melbourne, Parkville, VIC 3010, Australia.
| | | |
Collapse
|
37
|
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
|
38
|
Hippocampal neurogenesis, cognitive deficits and affective disorder in Huntington's disease. Neural Plast 2012; 2012:874387. [PMID: 22830053 PMCID: PMC3394391 DOI: 10.1155/2012/874387] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 05/13/2012] [Accepted: 05/14/2012] [Indexed: 02/07/2023] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by a tandem repeat expansion encoding a polyglutamine tract in the huntingtin protein. HD involves progressive psychiatric, cognitive, and motor symptoms, the selective pathogenesis of which remains to be mechanistically elucidated. There are a range of different brain regions, including the cerebral cortex and striatum, known to be affected in HD, with evidence for hippocampal dysfunction accumulating in recent years. In this review we will focus on hippocampal abnormalities, in particular, deficits of adult neurogenesis. We will discuss potential molecular mechanisms mediating disrupted hippocampal neurogenesis, and how this deficit of cellular plasticity may in turn contribute to specific cognitive and affective symptoms that are prominent in HD. The generation of transgenic animal models of HD has greatly facilitated our understanding of disease mechanisms at molecular, cellular, and systems levels. Transgenic HD mice have been found to show progressive behavioral changes, including affective, cognitive, and motor abnormalities. The discovery, in multiple transgenic lines of HD mice, that adult hippocampal neurogenesis and synaptic plasticity is disrupted, may help explain specific aspects of cognitive and affective dysfunction. Furthermore, these mouse models have provided insight into potential molecular mediators of adult neurogenesis deficits, such as disrupted serotonergic and neurotrophin signaling. Finally, a number of environmental and pharmacological interventions which are known to enhance adult hippocampal neurogenesis have been found to have beneficial affective and cognitive effects in mouse models, suggesting common molecular targets which may have therapeutic utility for HD and related diseases.
Collapse
|
39
|
Cellular neurochemical characterization and subcellular localization of phospholipase C β1 in rat brain. Neuroscience 2012; 222:239-68. [PMID: 22735577 DOI: 10.1016/j.neuroscience.2012.06.039] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 06/15/2012] [Accepted: 06/15/2012] [Indexed: 11/21/2022]
Abstract
The present study describes a complete and detailed neuroanatomical distribution map of the phospholipase C beta1 (PLCβ1) isoform along the adult rat neuraxis, and defines the phenotype of cells expressing PLCβ1, along with its subcellular localization in cortical neurons as assessed by double-immunofluorescence staining and confocal laser scanning. Immunohistochemical labeling revealed a considerable morphological heterogeneity among PLCβ1-positive cells in the cortex, even though there was a marked predominance of pyramidal morphologies. As an exception to the general non-matching distribution of GFAP and PLCβ1, a high degree of co-expression was observed in radial glia-like processes of the spinal cord white matter. In the somatosensory cortex, the proportion of GABAergic neurons co-stained with PLCβ1 was similar (around 2/3) in layers I, II-III, IV and VI, and considerably lower in layer V (around 2/5). Double immunofluorescence against PLCβ1 and nuclear speckle markers SC-35 and NeuN/Fox3 in isolated nuclei from the rat cortex showed a high overlap of both markers with PLCβ1 within the nuclear matrix. In contrast, there was no apparent co-localization with markers of the nuclear envelope and lamina. Finally, to assess whether the subcellular expression pattern of PLCβ1 involved specifically one of the two splice variants of PLCβ1, we carried out Western blot experiments in cortical subcellular fractions. Notably, PLCβ1a/1b ratios were statistically higher in the cytoplasm than in the nuclear and plasma membrane fractions. These results provide a deeper knowledge of the cellular distribution of the PLCβ1 isoform in different cell subtypes of the rat brain, and of its presence in the neuronal nuclear compartment.
Collapse
|
40
|
Effects of environmental manipulations in genetically targeted animal models of affective disorders. Neurobiol Dis 2012; 57:12-27. [PMID: 22525570 DOI: 10.1016/j.nbd.2012.04.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 04/02/2012] [Accepted: 04/06/2012] [Indexed: 12/31/2022] Open
Abstract
Mental illness is the leading cause of disability worldwide. We are only just beginning to reveal and comprehend the complex interaction that exists between the genetic makeup of an organism and the potential modifying effect of the environment in which it lives, and how this translates into mediating susceptibility to neurological and psychiatric conditions. The capacity to address this issue experimentally has been facilitated by the availability of rodent models which allow the precise manipulation of genetic and environmental factors. In this review, we discuss the valuable nature of animal models in furthering our understanding of the relationship between genetic and environmental factors in affective illnesses, such as anxiety and depressive disorders. We first highlight the behavioral impairments exhibited by genetically targeted animal models of affective disorders, and then provide a discussion of the underlying neurobiology, focusing on animal models that involve exposure to stress. This is followed by a review of recent studies that report of beneficial effects of environmental manipulations such as environmental enrichment and enhanced physical activity and discuss the likely mechanisms that mediate those benefits.
Collapse
|
41
|
Duveau V, Laustela S, Barth L, Gianolini F, Vogt KE, Keist R, Chandra D, Homanics GE, Rudolph U, Fritschy JM. Spatiotemporal specificity of GABAA receptor-mediated regulation of adult hippocampal neurogenesis. Eur J Neurosci 2011; 34:362-73. [PMID: 21722213 DOI: 10.1111/j.1460-9568.2011.07782.x] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
GABAergic transmission regulates adult neurogenesis by exerting negative feedback on cell proliferation and enabling dendrite formation and outgrowth. Further, GABAergic synapses target differentiating dentate gyrus granule cells prior to formation of glutamatergic connections. GABA(A) receptors (GABA(A) Rs) mediating tonic (extrasynaptic) and phasic (synaptic) transmission are molecularly and functionally distinct, but their specific role in regulating adult neurogenesis is unknown. Using global and single-cell targeted gene deletion of subunits contributing to the assembly of GABA(A) Rs mediating tonic (α4, δ) or phasic (α2) GABAergic transmission, we demonstrate here in the dentate gyrus of adult mice that GABA(A) Rs containing α4, but not δ, subunits mediate GABAergic effects on cell proliferation, initial migration and early dendritic development. In contrast, α2-GABA(A) Rs cell-autonomously signal to control positioning of newborn neurons and regulate late maturation of their dendritic tree. In particular, we observed pruning of distal dendrites in immature granule cells lacking the α2 subunit. This alteration could be prevented by pharmacological inhibition of thrombospondin signaling with chronic gabapentin treatment, shown previously to reduce glutamatergic synaptogenesis. These observations point to homeostatic regulation of inhibitory and excitatory inputs onto newborn granule cells under the control of α2-GABA(A) Rs. Taken together, the availability of distinct GABA(A) R subtypes provides a molecular mechanism endowing spatiotemporal specificity to GABAergic control of neuronal maturation in adult brain.
Collapse
Affiliation(s)
- Venceslas Duveau
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|