1
|
Denizci E, Altun G, Kaplan S. Morphological evidence for the potential protective effects of curcumin and Garcinia kola against diabetes in the rat hippocampus. Brain Res 2024; 1839:149020. [PMID: 38788929 DOI: 10.1016/j.brainres.2024.149020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/16/2024] [Accepted: 05/22/2024] [Indexed: 05/26/2024]
Abstract
This research investigated the effects of sciatic nerve transection and diabetes on the hippocampus, and the protective effects of Garcinia kola and curcumin. Thirty-five adults male Wistar albino rats were divided into five groups: a control group (Cont), a transected group (Sham group), a transected + diabetes mellitus group (DM), a transected + diabetes mellitus + Garcinia kola group (DM + GK), and a transected + DM + curcumin group (DM + Cur), each containing seven animals. The experimental diabetes model was created with the intraperitoneal injection of a single dose of streptozotocin. No procedure was applied to the Cont group, while sciatic nerve transection was performed on the other groups. Garcinia kola was administered to the rats in DM + GK, and curcumin to those in DM + Cur. Cardiac perfusion was performed at the end of the experimental period. Brain tissues were dissected for stereological, histopathological, and immunohistochemical evaluations. The volume ratios of hippocampal layers to the entire hippocampus volume were compared between the groups. Anti-S100, anti-caspase 3, and anti-SOX 2 antibodies were used for immunohistochemical analysis. No statistically significant difference was observed in the volume ratios of the four hippocampal layers. However, the volume ratio of the stratum lucidum was higher in the Sham, DM, and DM + Cur groups compared to the Cont group. While curcumin exhibited a protective effect on hippocampal tissue following diabetes induction, Garcinia kola had only a weak protective effect. Increased cell density and nuclear deterioration due to diabetes and nerve transection can be partially ameliorated by treatment with Garcinia kola and curcumin.
Collapse
Affiliation(s)
- Eda Denizci
- Department of Histology and Embryology, Ondokuz Mayıs University, Samsun 55139, Turkey
| | - Gamze Altun
- Department of Histology and Embryology, Ondokuz Mayıs University, Samsun 55139, Turkey
| | - Süleyman Kaplan
- Department of Histology and Embryology, Ondokuz Mayıs University, Samsun 55139, Turkey; Nelson Mandela African Institution of Science and Technology, Arusha, Tanzania.
| |
Collapse
|
2
|
Mhalhel K, Sicari M, Pansera L, Chen J, Levanti M, Diotel N, Rastegar S, Germanà A, Montalbano G. Zebrafish: A Model Deciphering the Impact of Flavonoids on Neurodegenerative Disorders. Cells 2023; 12:252. [PMID: 36672187 PMCID: PMC9856690 DOI: 10.3390/cells12020252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/17/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Over the past century, advances in biotechnology, biochemistry, and pharmacognosy have spotlighted flavonoids, polyphenolic secondary metabolites that have the ability to modulate many pathways involved in various biological mechanisms, including those involved in neuronal plasticity, learning, and memory. Moreover, flavonoids are known to impact the biological processes involved in developing neurodegenerative diseases, namely oxidative stress, neuroinflammation, and mitochondrial dysfunction. Thus, several flavonoids could be used as adjuvants to prevent and counteract neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. Zebrafish is an interesting model organism that can offer new opportunities to study the beneficial effects of flavonoids on neurodegenerative diseases. Indeed, the high genome homology of 70% to humans, the brain organization largely similar to the human brain as well as the similar neuroanatomical and neurochemical processes, and the high neurogenic activity maintained in the adult brain makes zebrafish a valuable model for the study of human neurodegenerative diseases and deciphering the impact of flavonoids on those disorders.
Collapse
Affiliation(s)
- Kamel Mhalhel
- Zebrafish Neuromorphology Lab., Department of Veterinary Sciences, University of Messina, Via Giovanni Palatucci snc, 98168 Messina, Italy
| | - Mirea Sicari
- Zebrafish Neuromorphology Lab., Department of Veterinary Sciences, University of Messina, Via Giovanni Palatucci snc, 98168 Messina, Italy
| | - Lidia Pansera
- Zebrafish Neuromorphology Lab., Department of Veterinary Sciences, University of Messina, Via Giovanni Palatucci snc, 98168 Messina, Italy
| | - Jincan Chen
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Campus North, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Maria Levanti
- Zebrafish Neuromorphology Lab., Department of Veterinary Sciences, University of Messina, Via Giovanni Palatucci snc, 98168 Messina, Italy
| | - Nicolas Diotel
- Université de la Réunion, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Plateforme CYROI, F-97490 Sainte-Clotilde, France
| | - Sepand Rastegar
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Campus North, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Antonino Germanà
- Zebrafish Neuromorphology Lab., Department of Veterinary Sciences, University of Messina, Via Giovanni Palatucci snc, 98168 Messina, Italy
| | - Giuseppe Montalbano
- Zebrafish Neuromorphology Lab., Department of Veterinary Sciences, University of Messina, Via Giovanni Palatucci snc, 98168 Messina, Italy
| |
Collapse
|
3
|
Liu C, Liu J, Gong H, Liu T, Li X, Fan X. Implication of Hippocampal Neurogenesis in Autism Spectrum Disorder: Pathogenesis and Therapeutic Implications. Curr Neuropharmacol 2023; 21:2266-2282. [PMID: 36545727 PMCID: PMC10556385 DOI: 10.2174/1570159x21666221220155455] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 12/24/2022] Open
Abstract
Autism spectrum disorder (ASD) is a cluster of heterogeneous neurodevelopmental conditions with atypical social communication and repetitive sensory-motor behaviors. The formation of new neurons from neural precursors in the hippocampus has been unequivocally demonstrated in the dentate gyrus of rodents and non-human primates. Accumulating evidence sheds light on how the deficits in the hippocampal neurogenesis may underlie some of the abnormal behavioral phenotypes in ASD. In this review, we describe the current evidence concerning pre-clinical and clinical studies supporting the significant role of hippocampal neurogenesis in ASD pathogenesis, discuss the possibility of improving hippocampal neurogenesis as a new strategy for treating ASD, and highlight the prospect of emerging pro-neurogenic therapies for ASD.
Collapse
Affiliation(s)
- Chuanqi Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
- Battalion 5 of Cadet Brigade, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jiayin Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
- Battalion 5 of Cadet Brigade, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hong Gong
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Tianyao Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xin Li
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
- Army 953 Hospital, Shigatse Branch of Xinqiao Hospital, Third Military Medical University (Army Medical University), Shigatse, China
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
4
|
Gao Y, Shen M, Gonzalez JC, Dong Q, Kannan S, Hoang JT, Eisinger BE, Pandey J, Javadi S, Chang Q, Wang D, Overstreet-Wadiche L, Zhao X. RGS6 Mediates Effects of Voluntary Running on Adult Hippocampal Neurogenesis. Cell Rep 2021; 32:107997. [PMID: 32755589 DOI: 10.1016/j.celrep.2020.107997] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/29/2020] [Accepted: 07/15/2020] [Indexed: 01/17/2023] Open
Abstract
Voluntary running enhances adult hippocampal neurogenesis, with consequences for hippocampal-dependent learning ability and mood regulation. However, the underlying mechanism remains unclear. Here, we show that voluntary running induces unique and dynamic gene expression changes specifically within the adult-born hippocampal neurons, with significant impact on genes involved in neuronal maturation and human diseases. We identify the regulator of G protein signaling 6 (RGS6) as a key factor that mediates running impact on adult-born neurons. RGS6 overexpression mimics the positive effects of voluntary running on morphological and physiological maturation of adult new neurons and reduced sensitivity of adult-born neurons to the inhibitory effect of GABAB (γ-Aminobutyric acid B) receptor activation. Knocking down RGS6 abolishes running-enhanced neuronal maturation and hippocampal neurogenesis-dependent learning and anxiolytic effect. Our study provides a data resource showing genome-wide intrinsic molecular changes in adult-born hippocampal neurons that contribute to voluntary running-induced neurogenesis.
Collapse
Affiliation(s)
- Yu Gao
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Minjie Shen
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jose Carlos Gonzalez
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Qiping Dong
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Sudharsan Kannan
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Johnson T Hoang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Brian E Eisinger
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jyotsna Pandey
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Sahar Javadi
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Qiang Chang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neurology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Daifeng Wang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | | | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
5
|
Calió ML, Henriques E, Siena A, Bertoncini CRA, Gil-Mohapel J, Rosenstock TR. Mitochondrial Dysfunction, Neurogenesis, and Epigenetics: Putative Implications for Amyotrophic Lateral Sclerosis Neurodegeneration and Treatment. Front Neurosci 2020; 14:679. [PMID: 32760239 PMCID: PMC7373761 DOI: 10.3389/fnins.2020.00679] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/03/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive and devastating multifactorial neurodegenerative disorder. Although the pathogenesis of ALS is still not completely understood, numerous studies suggest that mitochondrial deregulation may be implicated in its onset and progression. Interestingly, mitochondrial deregulation has also been associated with changes in neural stem cells (NSC) proliferation, differentiation, and migration. In this review, we highlight the importance of mitochondrial function for neurogenesis, and how both processes are correlated and may contribute to the pathogenesis of ALS; we have focused primarily on preclinical data from animal models of ALS, since to date no studies have evaluated this link using human samples. As there is currently no cure and no effective therapy to counteract ALS, we have also discussed how improving neurogenic function by epigenetic modulation could benefit ALS. In support of this hypothesis, changes in histone deacetylation can alter mitochondrial function, which in turn might ameliorate cellular proliferation as well as neuronal differentiation and migration. We propose that modulation of epigenetics, mitochondrial function, and neurogenesis might provide new hope for ALS patients, and studies exploring these new territories are warranted in the near future.
Collapse
Affiliation(s)
| | - Elisandra Henriques
- Department of Physiological Science, Santa Casa de São Paulo School of Medical Science, São Paulo, Brazil
| | - Amanda Siena
- Department of Physiological Science, Santa Casa de São Paulo School of Medical Science, São Paulo, Brazil
| | - Clélia Rejane Antonio Bertoncini
- CEDEME, Center of Development of Experimental Models for Medicine and Biology, Federal University of São Paulo, São Paulo, Brazil
| | - Joana Gil-Mohapel
- Division of Medical Sciences, Faculty of Medicine, University of Victoria and Island Medical Program, University of British Columbia, Victoria, BC, Canada
| | - Tatiana Rosado Rosenstock
- Department of Physiological Science, Santa Casa de São Paulo School of Medical Science, São Paulo, Brazil
| |
Collapse
|
6
|
Mahmoudi R, Ghareghani M, Zibara K, Tajali Ardakani M, Jand Y, Azari H, Nikbakht J, Ghanbari A. Alyssum homolocarpum seed oil (AHSO), containing natural alpha linolenic acid, stearic acid, myristic acid and β-sitosterol, increases proliferation and differentiation of neural stem cells in vitro. Altern Ther Health Med 2019; 19:113. [PMID: 31159797 PMCID: PMC6547481 DOI: 10.1186/s12906-019-2518-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 05/07/2019] [Indexed: 01/01/2023]
Abstract
Background Embryonic neural stem cells (eNSCs) are immature precursors of the central nervous system (CNS), with self-renewal and multipotential differentiation capacities. These are regulated by endogenous and exogenous factors such as alpha-linolenic acid (ALA), a plant-based essential omega-3 polyunsaturated fatty acid. Methods In this study, we investigated the effects of various concentrations of Alyssum homolocarpum seed oil (AHSO), containing natural ALA, stearic acid (SA), myristic acid (MA), and β-sitosterol, on proliferation and differentiation of eNSCs, in comparison to controls and to synthetic pure ALA. Results Treatment with natural AHSO (25 to 75 μM), similar to synthetic ALA, caused a significant ~ 2-fold increase in eNCSs viability, in comparison to controls. To confirm this proliferative activity, treatment of NSCs with 50 or 75 μM AHSO resulted in a significant increase in mRNA levels of notch1, hes-1 and Ki-67and NICD protein expression, in comparison to controls. Moreover, AHSO administration significantly increased the differentiation of eNSCs toward astrocytes (GFAP+) and oligodendrocytes (MBP+) in a dose dependent manner and was more potent than ALA, at similar concentrations, in comparison to controls. Indeed, only high concentrations of 100 μM AHSO, but not ALA, caused a significant increase in the frequency of neurons (β-III Tubulin+). Conclusion Our data demonstrated that AHSO, a rich source of ALA containing also other beneficial fatty acids, increased the proliferation and stimulated the differentiation of eNSCs. We suggest that AHSO’s effects are caused by β-sitosterol, SA and MA, present within this oil. AHSO could be used in diet to prevent neurodevelopmental syndromes, cognitive decline during aging, and various psychiatric disorders.
Collapse
|
7
|
Dhaliwal J, Kannangara TS, Vaculik M, Xue Y, Kumar KL, Maione A, Béïque JC, Shen J, Lagace DC. Adult hippocampal neurogenesis occurs in the absence of Presenilin 1 and Presenilin 2. Sci Rep 2018; 8:17931. [PMID: 30560948 PMCID: PMC6299003 DOI: 10.1038/s41598-018-36363-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 11/10/2018] [Indexed: 12/19/2022] Open
Abstract
Mutations in the presenilin genes (PS1 and PS2) are a major cause of familial-Alzheimer's disease (FAD). Presenilins regulate neurogenesis in the developing brain, with loss of PS1 inducing aberrant premature differentiation of neural progenitor cells, and additional loss of PS2 exacerbating this effect. It is unclear, however, whether presenilins are involved in adult neurogenesis, a process that may be impaired in Alzheimer's disease within the hippocampus. To investigate the requirement of presenilins in adult-generated dentate granule neurons, we examined adult neurogenesis in the PS2-/- adult brain and then employ a retroviral approach to ablate PS1 selectively in dividing progenitor cells of the PS2-/- adult brain. Surprisingly, the in vivo ablation of both presenilins resulted in no defects in the survival and differentiation of adult-generated neurons. There was also no change in the morphology or functional properties of the retroviral-labeled presenilin-null cells, as assessed by dendritic morphology and whole-cell electrophysiology analyses. Furthermore, while FACS analysis showed that stem and progenitor cells express presenilins, inactivation of presenilins from these cells, using a NestinCreERT2 inducible genetic approach, demonstrated no changes in the proliferation, survival, or differentiation of adult-generated cells. Therefore, unlike their significant role in neurogenesis during embryonic development, presenilins are not required for cell-intrinsic regulation of adult hippocampal neurogenesis.
Collapse
Affiliation(s)
- Jagroop Dhaliwal
- Department of Cellular and Molecular Medicine, Brain and Mind Research Institute, and Neuroscience Program, University of Ottawa, Ottawa, Ontario, K1H8M5, Canada
| | - Timal S Kannangara
- Department of Cellular and Molecular Medicine, Brain and Mind Research Institute, and Neuroscience Program, University of Ottawa, Ottawa, Ontario, K1H8M5, Canada
| | - Michael Vaculik
- Department of Cellular and Molecular Medicine, Brain and Mind Research Institute, and Neuroscience Program, University of Ottawa, Ottawa, Ontario, K1H8M5, Canada
| | - Yingben Xue
- Department of Cellular and Molecular Medicine, Brain and Mind Research Institute, and Neuroscience Program, University of Ottawa, Ottawa, Ontario, K1H8M5, Canada
| | - Keren L Kumar
- Department of Cellular and Molecular Medicine, Brain and Mind Research Institute, and Neuroscience Program, University of Ottawa, Ottawa, Ontario, K1H8M5, Canada
| | - Amanda Maione
- Department of Cellular and Molecular Medicine, Brain and Mind Research Institute, and Neuroscience Program, University of Ottawa, Ottawa, Ontario, K1H8M5, Canada
| | - Jean-Claude Béïque
- Department of Cellular and Molecular Medicine, Brain and Mind Research Institute, and Neuroscience Program, University of Ottawa, Ottawa, Ontario, K1H8M5, Canada
| | - Jie Shen
- Department of Neurology, Brigham and Women's Hospital and Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, 02115, USA
| | - Diane C Lagace
- Department of Cellular and Molecular Medicine, Brain and Mind Research Institute, and Neuroscience Program, University of Ottawa, Ottawa, Ontario, K1H8M5, Canada.
| |
Collapse
|
8
|
Ishimoto T, Masuo Y, Kato Y, Nakamichi N. Ergothioneine-induced neuronal differentiation is mediated through activation of S6K1 and neurotrophin 4/5-TrkB signaling in murine neural stem cells. Cell Signal 2018; 53:269-280. [PMID: 30359715 DOI: 10.1016/j.cellsig.2018.10.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 10/15/2018] [Accepted: 10/18/2018] [Indexed: 01/08/2023]
Abstract
The promotion of neurogenesis is considered to be an effective therapeutic strategy for neuropsychiatric disorders because impairment of neurogenesis is associated with the onset and progression of these disorders. We have previously demonstrated that orally ingested ergothioneine (ERGO), a naturally occurring antioxidant and hydrophilic amino acid, promotes neurogenesis in the hippocampal dentate gyrus (DG) with its abundant neural stem cells (NSCs) and exerts antidepressant-like effects in mice. Independent of its antioxidant activities, ERGO induces in cultured NSCs this differentiation through induction of the basic helix-loop-helix transcription factor Math1. However, the upstream signaling of Math1 in the mechanisms underlying ERGO-induced neuronal differentiation remains unclear. The purpose of the present study was to elucidate the upstream signaling with the aim of discovering novel targets for the treatment of neuropsychiatric disorders. We focused on neurotrophic factor signaling, as it is important for the promotion of neurogenesis and the induction of antidepressant effects. We also focused on the signaling of the mammalian target of rapamycin (mTOR) complex 1 (mTORC1), a known amino acid sensor, and the members of this signaling pathway, mTOR and p70 ribosomal protein S6 kinase 1 (S6K1). Exposure of cultured NSCs to ERGO significantly increased the expression of phosphorylated S6K1 (p-S6K1) at Thr389 in only 1 h, of phosphorylated mTOR (p-mTOR) in 6 h, and of the gene product of neurotrophin 4/5 (NT5) which activates tropomyosin receptor kinase B (TrkB) in 24 h. ERGO increased the population of βIII-tubulin-positive neurons, and this effect was suppressed by the inhibitors of S6K1 (PF4708671), mTORC1 (rapamycin), and TrkB (GNF5837). Oral administration of ERGO to mice significantly increased in the DG the expression of p-S6K1 at Thr389, the gene product of NT5, and phosphorylated TrkB but not that of p-mTOR. Thus, neuronal differentiation of NSCs induced by ERGO is mediated, at least in part, through phosphorylation of S6K1 at Thr389 and subsequent activation of TrkB signaling through the induction of NT5. Thus, S6K1 and NT5 might be promising target molecules for the treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Takahiro Ishimoto
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Yusuke Masuo
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Yukio Kato
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Noritaka Nakamichi
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan.
| |
Collapse
|
9
|
Zhang W, Wang W, Yu DX, Xiao Z, He Z. Application of nanodiagnostics and nanotherapy to CNS diseases. Nanomedicine (Lond) 2018; 13:2341-2371. [PMID: 30088440 DOI: 10.2217/nnm-2018-0163] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease, Parkinson's disease and stroke are the most common CNS diseases, all characterized by progressive cellular dysfunction and death in specific areas of the nervous system. Therapeutic development for these diseases has lagged behind other disease areas due to difficulties in early diagnosis, long disease courses and drug delivery challenges, not least due to the blood-brain barrier. Over recent decades, nanotechnology has been explored as a potential tool for the diagnosis, treatment and monitoring of CNS diseases. In this review, we describe the application of nanotechnology to common CNS diseases, highlighting disease pathogenesis and the underlying mechanisms and promising functional outcomes that make nanomaterials ideal candidates for early diagnosis and therapy. Moreover, we discuss the limitations of nanotechnology, and possible solutions.
Collapse
Affiliation(s)
- Weiyuan Zhang
- Yunnan Key Laboratory of Stem Cell & Regenerative Medicine, Institute of Molecular & Clinical Medicine, Kunming Medical University, Kunming 650500, PR China
| | - Wenyue Wang
- Department of Anatomy & Developmental Biology, Monash University, Clayton, 3800 Clayton, Melbourne 3800, Australia
| | - David X Yu
- Department of Anatomy & Developmental Biology, Monash University, Clayton, 3800 Clayton, Melbourne 3800, Australia
| | - Zhicheng Xiao
- Department of Anatomy & Developmental Biology, Monash University, Clayton, 3800 Clayton, Melbourne 3800, Australia
| | - Zhiyong He
- Yunnan Key Laboratory of Stem Cell & Regenerative Medicine, Institute of Molecular & Clinical Medicine, Kunming Medical University, Kunming 650500, PR China.,Department of Anatomy & Developmental Biology, Monash University, Clayton, 3800 Clayton, Melbourne 3800, Australia
| |
Collapse
|
10
|
Jeon SW, Kim YK. The role of neuroinflammation and neurovascular dysfunction in major depressive disorder. J Inflamm Res 2018; 11:179-192. [PMID: 29773951 PMCID: PMC5947107 DOI: 10.2147/jir.s141033] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Although depression has generally been explained with monoamine theory, it is far more multifactorial, and therapies that address the disease’s pathway have not been developed. In this context, an understanding of neuroinflammation and neurovascular dysfunction would enable a more comprehensive approach to depression. Inflammation is in a sense a type of allostatic load involving the immune, endocrine, and nervous systems. Neuroinflammation is involved in the pathophysiology of depression by increasing proinflammatory cytokines, activating the hypothalamus–pituitary–adrenal axis, increasing glucocorticoid resistance, and affecting serotonin synthesis and metabolism, neuronal apoptosis and neurogenesis, and neuroplasticity. In future, identifying the subtypes of depression with increased vulnerability to inflammation and testing the effects of inflammatory modulating agents in these patient groups through clinical trials will lead to more concrete conclusions on the matter. The vascular depression hypothesis is supported by evidence for the association between vascular disease and late-onset depression and between ischemic brain lesions and distinctive depressive symptoms. Vascular depression may be the entity most suitable for studies of the mechanisms of depression. Pharmacotherapies used in the prevention and treatment of cerebrovascular disease may help prevent vascular depression. In future, developments in structural and functional imaging, electrophysiology, chronobiology, and genetics will reveal the association between depression and brain lesions. This article aims to give a general review of the existing issues examined in the literature pertaining to depression-related neuroinflammatory and vascular functions, related pathophysiology, applicability to depression treatment, and directions for future research.
Collapse
Affiliation(s)
- Sang Won Jeon
- Department of Psychiatry, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul
| | - Yong-Ku Kim
- Department of Psychiatry, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, South Korea
| |
Collapse
|
11
|
Barbieri R, Contestabile A, Ciardo MG, Forte N, Marte A, Baldelli P, Benfenati F, Onofri F. Synapsin I and Synapsin II regulate neurogenesis in the dentate gyrus of adult mice. Oncotarget 2018; 9:18760-18774. [PMID: 29721159 PMCID: PMC5922353 DOI: 10.18632/oncotarget.24655] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 02/25/2018] [Indexed: 01/23/2023] Open
Abstract
Adult neurogenesis is emerging as an important player in brain functions and homeostasis, while impaired or altered adult neurogenesis has been associated with a number of neuropsychiatric diseases, such as depression and epilepsy. Here we investigated the possibility that synapsins (Syns) I and II, beyond their known functions in developing and mature neurons, also play a role in adult neurogenesis. We performed a systematic evaluation of the distinct stages of neurogenesis in the hippocampal dentate gyrus of Syn I and Syn II knockout (KO) mice, before (2-months-old) and after (6-months-old) the appearance of the epileptic phenotype. We found that Syns I and II play an important role in the regulation of adult neurogenesis. In juvenile mice, Syn II deletion was associated with a specific decrease in the proliferation of neuronal progenitors, whereas Syn I deletion impaired the survival of newborn neurons. These defects were reverted after the appearance of the epileptic phenotype, with Syn I KO and Syn II KO mice exhibiting significant increases in survival and proliferation, respectively. Interestingly, long-term potentiation dependent on newborn neurons was present in both juvenile Syn mutants while, at later ages, it was only preserved in Syn II KO mice that also displayed an increased expression of brain-derived neurotrophic factor. This study suggests that Syns I and II play a role in adult neurogenesis and the defects in neurogenesis associated with Syn deletion may contribute to the alterations of cognitive functions observed in Syn-deficient mice.
Collapse
Affiliation(s)
- Raffaella Barbieri
- Department of Experimental Medicine, University of Genova, 16132, Genova, Italy
| | - Andrea Contestabile
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, 16163, Genova, Italy
| | - Maria Grazia Ciardo
- Department of Experimental Medicine, University of Genova, 16132, Genova, Italy
| | - Nicola Forte
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132, Genova, Italy
| | - Antonella Marte
- Department of Experimental Medicine, University of Genova, 16132, Genova, Italy
| | - Pietro Baldelli
- Department of Experimental Medicine, University of Genova, 16132, Genova, Italy.,Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132, Genova, Italy
| | - Fabio Benfenati
- Department of Experimental Medicine, University of Genova, 16132, Genova, Italy.,Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132, Genova, Italy
| | - Franco Onofri
- Department of Experimental Medicine, University of Genova, 16132, Genova, Italy
| |
Collapse
|
12
|
Abstract
BACKGROUND The microtubule-associated protein Tau plays a role in neurodegeneration as well as neurogenesis. Previous work has shown that the expression of the pro-aggregant mutant Tau repeat domain causes strong aggregation and pronounced neuronal loss in the hippocampus whereas the anti-aggregant form has no deleterious effects. These two proteins differ mainly in their propensity to form ß structure and hence to aggregate. METHODS To elucidate the basis of these contrasting effects, we analyzed organotypic hippocampal slice cultures (OHSCs) from transgenic mice expressing the repeat domain (RD) of Tau with the anti-aggregant mutation (TauRDΔKPP) and compared them with slices containing pro-aggregant TauRDΔK. Transgene expression in the hippocampus was monitored via a sensitive bioluminescence reporter gene assay (luciferase). RESULTS The expression of the anti-aggregant TauRDΔKPP leads to a larger volume of the hippocampus at a young age due to enhanced neurogenesis, resulting in an increase in neuronal number. There were no signs of activation of microglia and astrocytes, indicating the absence of an inflammatory reaction. Investigation of signaling pathways showed that Wnt-5a was strongly decreased whereas Wnt3 was increased. A pronounced increase in hippocampal stem cell proliferation (seen by BrdU) was observed as early as P8, in the CA regions where neurogenesis is normally not observed. The increase in neurons persisted up to 16 months of age. CONCLUSION The data suggest that the expression of anti-aggregant TauRDΔKPP enhances hippocampal neurogenesis mediated by the canonical Wnt signaling pathway, without an inflammatory reaction. This study points to a role of tau in brain development and neurogenesis, in contrast to its detrimental role in neurodegeneration at later age.
Collapse
|
13
|
Navarro-Sanchis C, Brock O, Winsky-Sommerer R, Thuret S. Modulation of Adult Hippocampal Neurogenesis by Sleep: Impact on Mental Health. Front Neural Circuits 2017; 11:74. [PMID: 29075182 PMCID: PMC5643465 DOI: 10.3389/fncir.2017.00074] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 09/26/2017] [Indexed: 12/27/2022] Open
Abstract
The process of neurogenesis has been demonstrated to occur throughout life in the subgranular zone (SGZ) of the hippocampal dentate gyrus of several mammals, including humans. The basal rate of adult hippocampal neurogenesis can be altered by lifestyle and environmental factors. In this perspective review, the evidence for sleep as a modulator of adult hippocampal neurogenesis is first summarized. Following this, the impacts of sleep and sleep disturbances on hippocampal-dependent functions, including learning and memory, and depression are critically evaluated. Finally, we postulate that the effects of sleep on hippocampal-dependent functions may possibly be mediated by a change in adult hippocampal neurogenesis. This could provide a route to new treatments for cognitive impairments and psychiatric disorders.
Collapse
Affiliation(s)
- Cristina Navarro-Sanchis
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Olivier Brock
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Raphaelle Winsky-Sommerer
- Surrey Sleep Research Centre, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford, United Kingdom
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
14
|
Galán L, Gómez-Pinedo U, Guerrero A, García-Verdugo JM, Matías-Guiu J. Amyotrophic lateral sclerosis modifies progenitor neural proliferation in adult classic neurogenic brain niches. BMC Neurol 2017; 17:173. [PMID: 28874134 PMCID: PMC5585932 DOI: 10.1186/s12883-017-0956-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/30/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Adult neurogenesis persists through life at least in classic neurogenic niches. Neurogenesis has been previously described as reduced in neurodegenerative diseases. There is not much knowledge about is adult neurogenesis is or not modified in amyotrophy lateral sclerosis (ALS). All previous publications has studied the ALS SOD1 (superoxide dismutase) transgenic mouse model. The purpose of this study is to examine the process of adult neurogenesis in classic niches (subventricular zone [SVZ] and subgranular zone [SGZ] of the dentate gyrus) in patients with amyotrophic lateral sclerosis (ALS), both with (ALS-FTD) and without associated frontotemporal dementia (FTD). METHODS We studied 9 autopsies of patients with ALS (including 2 with ALS-FTD) and 4 controls. ALS was confirmed histologically. Studies of the SVZ and SGZ were conducted using markers of proliferation (Ki-67, PCNA), of pluripotent neural progenitor cells (GFAPδ), neuroblasts (PSA-NCAM, DCX, TUJ1), and an astrocyte marker (GFAP). Results were analyzed with non-parametric tests. We then studied correlations between the different markers and the percentage of phosphorylated TDP-43 (pTDP-43). RESULTS We observed a statistically significant increase in proliferation in the SVZ in all patients with ALS. While this increase was more marked in ALS forms associated with dementia, the small sample size does not permit a statistical subgroup analysis. In contrast, proliferation in the SGZ was decreased in all patients. These alterations showed a positive and direct correlation with the percentage of pTDP-43 in the SVZ, and a negative, exponential correlation with that percentage in the SGZ. CONCLUSIONS We observed alterations of the proliferation of neural progenitor in classic adult neurogenic niches in patients with ALS. The 2 neurogenic niches exhibited opposite changes such that proliferation increased in the SVZ and decreased in the SGZ.
Collapse
Affiliation(s)
- Lucía Galán
- Amyotrophic Lateral Sclerosis Unit, Department of Neurology, Hospital Clínico San Carlos, Calle Profesor Martín Lagos s/n, 28040 Madrid, Spain
| | | | - Antonio Guerrero
- Amyotrophic Lateral Sclerosis Unit, Department of Neurology, Hospital Clínico San Carlos, Calle Profesor Martín Lagos s/n, 28040 Madrid, Spain
| | - Jose Manuel García-Verdugo
- Cavanilles Institute of Biodiversity and Evolutionary Biology, Comparative Neurobiology Unit, Universidad de Valencia, Paterna, Spain
| | - Jorge Matías-Guiu
- Institute of Neurosciences, Hospital Clínico San Carlos, Madrid, Spain
| |
Collapse
|
15
|
Ihunwo AO, Tembo LH, Dzamalala C. The dynamics of adult neurogenesis in human hippocampus. Neural Regen Res 2016; 11:1869-1883. [PMID: 28197172 PMCID: PMC5270414 DOI: 10.4103/1673-5374.195278] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2016] [Indexed: 02/06/2023] Open
Abstract
The phenomenon of adult neurogenesis is now an accepted occurrence in mammals and also in humans. At least two discrete places house stem cells for generation of neurons in adult brain. These are olfactory system and the hippocampus. In animals, newly generated neurons have been directly or indirectly demonstrated to generate a significant amount of new neurons to have a functional role. However, the data in humans on the extent of this process is still scanty and such as difficult to comprehend its functional role in humans. This paper explores the available data on as extent of adult hippocampal neurogenesis in humans and makes comparison to animal data.
Collapse
Affiliation(s)
- Amadi O. Ihunwo
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Lackson H. Tembo
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Charles Dzamalala
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
16
|
The effects of hormones and physical exercise on hippocampal structural plasticity. Front Neuroendocrinol 2016; 41:23-43. [PMID: 26989000 DOI: 10.1016/j.yfrne.2016.03.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 03/02/2016] [Accepted: 03/08/2016] [Indexed: 01/22/2023]
Abstract
The hippocampus plays an integral role in certain aspects of cognition. Hippocampal structural plasticity and in particular adult hippocampal neurogenesis can be influenced by several intrinsic and extrinsic factors. Here we review how hormones (i.e., intrinsic modulators) and physical exercise (i.e., an extrinsic modulator) can differentially modulate hippocampal plasticity in general and adult hippocampal neurogenesis in particular. Specifically, we provide an overview of the effects of sex hormones, stress hormones, and metabolic hormones on hippocampal structural plasticity and adult hippocampal neurogenesis. In addition, we also discuss how physical exercise modulates these forms of hippocampal plasticity, giving particular emphasis on how this modulation can be affected by variables such as exercise regime, duration, and intensity. Understanding the neurobiological mechanisms underlying the modulation of hippocampal structural plasticity by intrinsic and extrinsic factors will impact the design of new therapeutic approaches aimed at restoring hippocampal plasticity following brain injury or neurodegeneration.
Collapse
|
17
|
Herbert J, Lucassen PJ. Depression as a risk factor for Alzheimer's disease: Genes, steroids, cytokines and neurogenesis - What do we need to know? Front Neuroendocrinol 2016; 41:153-71. [PMID: 26746105 DOI: 10.1016/j.yfrne.2015.12.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/23/2015] [Accepted: 12/27/2015] [Indexed: 01/18/2023]
Abstract
Depression (MDD) is prodromal to, and a component of, Alzheimer's disease (AD): it may also be a trigger for incipient AD. MDD is not a unitary disorder, so there may be particular subtypes of early life MDD that pose independent high risks for later AD, though the identification of these subtypes is problematical. There may either be a common pathological event underlying both MDD and AD, or MDD may sensitize the brain to a second event ('hit') that precipitates AD. MDD may also accelerate brain ageing, including altered DNA methylation, increased cortisol but decreasing DHEA and thus the risk for AD. So far, genes predicting AD (e.g. APOEε4) are not risk factors for MDD, and those implicated in MDD (e.g. SLC6A4) are not risks for AD, so a common genetic predisposition looks unlikely. There is as yet no strong indication that an epigenetic event occurs during some forms of MDD that predisposes to later AD, though the evidence is limited. Glucocorticoids (GCs) are disturbed in some cases of MDD and in AD. GCs have marked degenerative actions on the hippocampus, a site of early β-amyloid deposition, and rare genetic variants of GC-regulating enzymes (e.g. 11β-HSD) predispose to AD. GCs also inhibit hippocampal neurogenesis and plasticity, and thus episodic memory, a core symptom of AD. Disordered GCs in MDD may inhibit neurogenesis, but the contribution of diminished neurogenesis to the onset or progression of AD is still debated. GCs and cytokines also reduce BDNF, implicated in both MDD and AD and hippocampal neurogenesis, reinforcing the notion that those cases of MDD with disordered GCs may be a risk for AD. Cytokines, including IL1β, IL6 and TNFα, are increased in the blood in some cases of MDD. They also reduce hippocampal neurogenesis, and increased cytokines are a known risk for later AD. Inflammatory changes occur in both MDD and AD (e.g. raised CRP, TNFα). Both cytokines and GCs can have pro-inflammatory actions in the brain. Inflammation (e.g. microglial activation) may be a common link, but this has not been systematically investigated. We lack substantial, rigorous and comprehensive follow-up studies to better identify possible subtypes of MDD that may represent a major predictor for later AD. This would enable specific interventions during critical episodes of these subtypes of MDD that should reduce this substantial risk.
Collapse
Affiliation(s)
- Joe Herbert
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, UK.
| | - Paul J Lucassen
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, The Netherlands
| |
Collapse
|
18
|
Capilla-Gonzalez V, Bonsu JM, Redmond KJ, Garcia-Verdugo JM, Quiñones-Hinojosa A. Implications of irradiating the subventricular zone stem cell niche. Stem Cell Res 2016; 16:387-96. [PMID: 26921873 PMCID: PMC8442998 DOI: 10.1016/j.scr.2016.02.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 01/10/2016] [Accepted: 02/14/2016] [Indexed: 01/19/2023] Open
Abstract
Radiation therapy is a standard treatment for brain tumor patients. However, it comes with side effects, such as neurological deficits. While likely multi-factorial, the effect may in part be associated with the impact of radiation on the neurogenic niches. In the adult mammalian brain, the neurogenic niches are localized in the subventricular zone (SVZ) of the lateral ventricles and the dentate gyrus of the hippocampus, where the neural stem cells (NSCs) reside. Several reports showed that radiation produces a drastic decrease in the proliferative capacity of these regions, which is related to functional decline. In particular, radiation to the SVZ led to a reduced long-term olfactory memory and a reduced capacity to respond to brain damage in animal models, as well as compromised tumor outcomes in patients. By contrast, other studies in humans suggested that increased radiation dose to the SVZ may be associated with longer progression-free survival in patients with high-grade glioma. In this review, we summarize the cellular and functional effects of irradiating the SVZ niche. In particular, we review the pros and cons of using radiation during brain tumor treatment, discussing the complex relationship between radiation dose to the SVZ and both tumor control and toxicity.
Collapse
Affiliation(s)
- Vivian Capilla-Gonzalez
- Department of Neurosurgery and Oncology, Johns Hopkins University, Baltimore, MD 21231, USA; Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Seville 41092, Spain
| | - Janice M Bonsu
- Department of Neurosurgery and Oncology, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Kristin J Redmond
- Department of Radiation Oncology & Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Jose Manuel Garcia-Verdugo
- Laboratory of Comparative Neurobiology, Instituto Cavanilles de Biodiversidad y Biologia Evolutiva, University of Valencia, CIBERNED, Paterna 46980, Valencia, Spain
| | | |
Collapse
|
19
|
Komuro Y, Xu G, Bhaskar K, Lamb BT. Human tau expression reduces adult neurogenesis in a mouse model of tauopathy. Neurobiol Aging 2015; 36:2034-42. [PMID: 25863528 DOI: 10.1016/j.neurobiolaging.2015.03.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 02/24/2015] [Accepted: 03/03/2015] [Indexed: 11/29/2022]
Abstract
Accumulation of hyperphosphorylated and aggregated microtubule-associated protein tau (MAPT) is a central feature of a class of neurodegenerative diseases termed tauopathies. Notably, there is increasing evidence that tauopathies, including Alzheimer's disease, are also characterized by a reduction in neurogenesis, the birth of adult neurons. However, the exact relationship between hyperphosphorylation and aggregation of MAPT and neurogenic deficits remains unclear, including whether this is an early- or late-stage disease marker. In the present study, we used the genomic-based hTau mouse model of tauopathy to examine the temporal and spatial regulation of adult neurogenesis during the course of the disease. Surprisingly, hTau mice exhibited reductions in adult neurogenesis in 2 different brain regions by as early as 2 months of age, before the development of robust MAPT pathology in this model. This reduction was found to be due to reduced proliferation and not because of enhanced apoptosis in the hippocampus. At these same time points, hTau mice also exhibited altered MAPT phosphorylation with neurogenic precursors. To examine whether the effects of MAPT on neurogenesis were cell autonomous, neurospheres prepared from hTau animals were examined in vitro, revealing a growth deficit when compared with non-transgenic neurosphere cultures. Taken together, these studies provide evidence that altered adult neurogenesis is a robust and early marker of altered, cell-autonomous function of MAPT in the hTau mouse mode of tauopathy and that altered adult neurogenesis should be examined as a potential marker and therapeutic target for human tauopathies.
Collapse
Affiliation(s)
- Yutaro Komuro
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, NC30, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Guixiang Xu
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, NC30, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Kiran Bhaskar
- Department of Molecular Genetics and Microbiology, MIND Institute, University of New Mexico, MSC08 4660, 1 University of New Mexico, Albuquerque, NM, USA
| | - Bruce T Lamb
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, NC30, 9500 Euclid Avenue, Cleveland, OH, USA.
| |
Collapse
|
20
|
Hippocampal volume and shape in pure subcortical vascular dementia. Neurobiol Aging 2015; 36:485-91. [DOI: 10.1016/j.neurobiolaging.2014.08.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 08/01/2014] [Accepted: 08/07/2014] [Indexed: 01/18/2023]
|
21
|
Kokošová N, Tomášová L, Kisková T, Šmajda B. Neuronal analysis and behaviour in prenatally gamma-irradiated rats. Cell Mol Neurobiol 2015; 35:45-55. [PMID: 25537960 DOI: 10.1007/s10571-014-0144-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 11/19/2014] [Indexed: 01/09/2023]
Abstract
The intrauterinal development in mammals represents a very sensitive period of life in relation to many environmental factors, including ionizing radiation (IR). The developing nervous system is particularly vulnerable to IR, and the consequences of exposure are of importance because of its potential health risks. The aim of our work was to assess whether prenatal irradiation of rats on the 17th day of embryonic development with a dose of 1 Gy would affect the formation of new cells and the number of mature neurons in the hippocampus and the selected forms of behaviour in the postnatal period. Male progeny of irradiated and control females was tested at ages of 3 weeks, 2 and 3 months. The number of mitotically active cells in the gyrus dentatus (GD) of the hippocampus was significantly reduced in irradiated rats aged 3 weeks. In irradiated rats aged 2 months, a significant reduction of mature neurons in CA1 area and in GD of the hippocampus was observed. The IR negatively influenced the spatial memory in Morris water maze, significantly decreased the exploratory behaviour and increased the anxiety-like behaviour in elevated plus-maze in rats aged 2 months. No significant differences were observed in animals aged 3 months compared with controls of the same age. A significant correlation between the number of mature neurons in the hilus and of the cognitive performances was found. Our results show that a low dose of radiation applied during the sensitive phase of brain development can influence the level of neurogenesis in the subgranular zone of GD and cause an impairment of the postnatal development of mental functions.
Collapse
Affiliation(s)
- Natália Kokošová
- Department of Animal Physiology, Faculty of Science, Institute of Biology and Ecology, P.J.Šafárik University, Šrobárova 2, 040 01, Košice, Slovak Republic,
| | | | | | | |
Collapse
|
22
|
Physical exercise-induced adult neurogenesis: a good strategy to prevent cognitive decline in neurodegenerative diseases? BIOMED RESEARCH INTERNATIONAL 2014; 2014:403120. [PMID: 24818140 PMCID: PMC4000963 DOI: 10.1155/2014/403120] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 02/16/2014] [Accepted: 02/16/2014] [Indexed: 01/19/2023]
Abstract
Cumulative evidence has indicated that there is an important role for adult hippocampal neurogenesis in cognitive function. With the increasing prevalence of cognitive decline associated with neurodegenerative diseases among the ageing population, physical exercise, a potent enhancer of adult hippocampal neurogenesis, has emerged as a potential preventative strategy/treatment to reduce cognitive decline. Here we review the functional role of adult hippocampal neurogenesis in learning and memory, and how this form of structural plasticity is altered in neurodegenerative diseases known to involve cognitive impairment. We further discuss how physical exercise may contribute to cognitive improvement in the ageing brain by preserving adult neurogenesis, and review the recent approaches for measuring changes in neurogenesis in the live human brain.
Collapse
|
23
|
Abstract
The ubiquitous gaseous signaling molecule nitric oxide participates in the regulation of a variety of physiological and pathological processes, including adult neurogenesis. Adult neurogenesis, or the generation of new neurons in the adult brain, is a restricted event confined to areas with neurogenic capability. Although nitric oxide has been shown to mediate conflicting effects on adult neurogenesis, which may be partly explained by its unique characteristics, more studies are required in order to fully comprehend and appreciate the mechanisms involved. Neuropeptide Y, a neurotransmitter shown to be an important regulator of adult hippocampal neurogenesis, acts through intracellular nitric oxide to induce an increase in neural progenitor cell proliferation.
Collapse
|
24
|
Koutseff A, Mittelhaeuser C, Essabri K, Auwerx J, Meziane H. Impact of the apolipoprotein E polymorphism, age and sex on neurogenesis in mice: Pathophysiological relevance for Alzheimer's disease? Brain Res 2014; 1542:32-40. [DOI: 10.1016/j.brainres.2013.10.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 09/26/2013] [Accepted: 10/03/2013] [Indexed: 01/18/2023]
|
25
|
Marlatt MW, Potter MC, Bayer TA, van Praag H, Lucassen PJ. Prolonged running, not fluoxetine treatment, increases neurogenesis, but does not alter neuropathology, in the 3xTg mouse model of Alzheimer's disease. Curr Top Behav Neurosci 2013; 15:313-40. [PMID: 23670818 DOI: 10.1007/7854_2012_237] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Reductions in adult neurogenesis have been documented in the original 3xTg mouse model of Alzheimer's disease (AD), notably occurring at the same age when spatial memory deficits and amyloid plaque pathology appeared. As this suggested reduced neurogenesis was associated with behavioral deficits, we tested whether activity and pharmacological stimulation could prevent memory deficits and modify neurogenesis and/or neuropathology in the 3xTg model backcrossed to the C57Bl/6 strain. We chronically administered the antidepressant fluoxetine to one group of mice, allowed access to a running wheel in another, and combined both treatments in a third cohort. All treatments lasted for 11 months. The female 3xTg mice failed to exhibit any deficits in spatial learning and memory as measured in the Morris water maze, indicating that when backcrossed to the C57Bl/6 strain, the 3xTg mice lost the behavioral phenotype that was present in the original 3xTg mouse maintained on a hybrid background. Despite this, the backcrossed 3xTg mice expressed prominent intraneuronal amyloid beta (Aβ) levels in the cortex and amygdala, with lower levels in the CA1 area of the hippocampus. In the combined cohort, fluoxetine treatment interfered with exercise and reduced the total distance run. The extent of Aβ neuropathology, the tau accumulations, or BDNF levels, were not altered by prolonged exercise. Thus, neuropathology was present but not paralleled by spatial memory deficits in the backcrossed 3xTg mouse model of AD. Prolonged exercise for 11 months did improve the long-term survival of newborn neurons generated during middle-age, whereas fluoxetine had no effect. We further review and discuss the relevant literature in this respect.
Collapse
Affiliation(s)
- Michael W Marlatt
- Swammerdam Institute for Life Science-Center for Neuroscience, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
26
|
Iascone DM, Padidam S, Pyfer MS, Zhang X, Zhao L, Chin J. Impairments in neurogenesis are not tightly linked to depressive behavior in a transgenic mouse model of Alzheimer's disease. PLoS One 2013; 8:e79651. [PMID: 24244537 PMCID: PMC3828273 DOI: 10.1371/journal.pone.0079651] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 10/04/2013] [Indexed: 02/03/2023] Open
Abstract
Alzheimer's disease (AD), the most common cause of dementia, is also associated with depression. Although the precise mechanisms that lead to depression in AD are unknown, the impairments in adult hippocampal neurogenesis observed in AD may play a role. Adult-born neurons play a critical role in regulating both cognition and mood, and reduced hippocampal neurogenesis is associated with depression in other neurological disorders. To assess the relationship between Alzheimer's disease, neurogenesis, and depression, we studied human amyloid precursor protein (hAPP) transgenic mice, a well-characterized model of AD. We report that reductions in hippocampal neurogenesis are evident early in disease progression in hAPP mice, but a mild depressive phenotype manifests only in later stages of disease. We found that hAPP mice exhibited a reduction in BrdU-positive cells in the subgranular zone of the dentate gyrus in the hippocampus, as well as a reduction in doublecortin-expressing cells, relative to nontransgenic controls at 5-7 months of age. These alterations in neurogenesis appeared to worsen with age, as the magnitude of reduction in doublecortin-expressing cells was greater in hAPP mice at 13-15 months of age. Only 13-15 month old hAPP mice exhibited depressive behavior in the tail suspension test. However, mice at both age groups exhibited deficits in spatial memory, which was observed in the Morris water maze test for hippocampus-dependent memory. These findings indicate that neurogenesis impairments are accompanied by cognitive deficits, but are not tightly linked to depressive behavior in hAPP mice.
Collapse
Affiliation(s)
- Daniel M. Iascone
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Sneha Padidam
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Mark S. Pyfer
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Xiaohong Zhang
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Lijuan Zhao
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Jeannie Chin
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
27
|
Bielefeld P, van Vliet EA, Gorter JA, Lucassen PJ, Fitzsimons CP. Different subsets of newborn granule cells: a possible role in epileptogenesis? Eur J Neurosci 2013; 39:1-11. [DOI: 10.1111/ejn.12387] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Revised: 09/03/2013] [Accepted: 09/10/2013] [Indexed: 12/27/2022]
Affiliation(s)
- Pascal Bielefeld
- Center for Neuroscience; Swammerdam Institute for Life Sciences; University of Amsterdam; Science Park 904 1098 XH Amsterdam The Netherlands
| | - Erwin A. van Vliet
- Center for Neuroscience; Swammerdam Institute for Life Sciences; University of Amsterdam; Science Park 904 1098 XH Amsterdam The Netherlands
- Epilepsy Institute in The Netherlands Foundation (Stichting Epilepsie Instellingen Nederland SEIN); Heemstede The Netherlands
| | - Jan A. Gorter
- Center for Neuroscience; Swammerdam Institute for Life Sciences; University of Amsterdam; Science Park 904 1098 XH Amsterdam The Netherlands
| | - Paul J. Lucassen
- Center for Neuroscience; Swammerdam Institute for Life Sciences; University of Amsterdam; Science Park 904 1098 XH Amsterdam The Netherlands
| | - Carlos P. Fitzsimons
- Center for Neuroscience; Swammerdam Institute for Life Sciences; University of Amsterdam; Science Park 904 1098 XH Amsterdam The Netherlands
| |
Collapse
|
28
|
Oomen CA, Hvoslef-Eide M, Heath CJ, Mar AC, Horner AE, Bussey TJ, Saksida LM. The touchscreen operant platform for testing working memory and pattern separation in rats and mice. Nat Protoc 2013; 8:2006-21. [PMID: 24051961 PMCID: PMC3982138 DOI: 10.1038/nprot.2013.124] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The automated touchscreen operant chamber for rats and mice allows for the assessment of multiple cognitive domains within the same testing environment. This protocol presents the location discrimination (LD) task and the trial-unique delayed nonmatching-to-location (TUNL) task, which both assess memory for location. During these tasks, animals are trained to a predefined criterion during ∼20-40 daily sessions. In LD sessions, touching the same location on the screen is rewarded on consecutive trials, followed by a reversal of location-reward contingencies. TUNL, a working memory task, requires animals to 'nonmatch' to a sample location after a delay. In both the LD and TUNL tasks, spatial similarity can be varied, allowing assessment of pattern separation ability, a function that is thought to be performed by the dentate gyrus (DG). These tasks are therefore particularly useful in animal models of hippocampal, and specifically DG, function, but they additionally permit discernment of changes in pattern separation from those in working memory.
Collapse
|
29
|
Sutherland GT, Sheahan PJ, Matthews J, Dennis CVP, Sheedy DS, McCrossin T, Curtis MA, Kril JJ. The effects of chronic alcoholism on cell proliferation in the human brain. Exp Neurol 2013; 247:9-18. [PMID: 23541433 PMCID: PMC4709019 DOI: 10.1016/j.expneurol.2013.03.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 03/18/2013] [Accepted: 03/20/2013] [Indexed: 01/19/2023]
Abstract
Neurogenesis continues in the human subventricular zone and to a lesser extent in the hippocampal subgranular zone throughout life. Subventricular zone-derived neuroblasts migrate to the olfactory bulb where survivors become integrated as interneurons and are postulated to contribute to odor discrimination. Adult neurogenesis is dysregulated in many neurological, neurovascular and neurodegenerative diseases. Alcohol abuse can result in a neurodegenerative condition called alcohol-related brain damage. Alcohol-related brain damage manifests clinically as cognitive dysfunction and the loss of smell sensation (hyposmia) and pathologically as generalized white matter atrophy and focal neuronal loss. The exact mechanism linking chronic alcohol intoxication with alcohol-related brain damage remains largely unknown but rodent models suggest that decreased neurogenesis is an important component. We investigated this idea by comparing proliferative events in the subventricular zone and olfactory bulb of a well-characterized cohort of 15 chronic alcoholics and 16 age-matched controls. In contrast to the findings in animal models there was no difference in the number of proliferative cell nuclear antigen-positive cells in the subventricular zone of alcoholics (mean±SD=28.7±20.0) and controls (27.6±18.9, p=1.0). There were also no differences in either the total (p=0.89) or proliferative cells (p=0.98) in the granular cell layer of the olfactory bulb. Our findings show that chronic alcohol consumption does not affect cell proliferation in the human SVZ or olfactory bulb. In fact only microglial proliferation could be demonstrated in the latter. Therefore neurogenic deficits are unlikely to contribute to hyposmia in chronic alcoholics.
Collapse
Affiliation(s)
- G T Sutherland
- Discipline of Pathology, Sydney Medical School, The University of Sydney, NSW 2006, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Aβ increases neural stem cell activity in senescence-accelerated SAMP8 mice. Neurobiol Aging 2013; 34:2623-38. [PMID: 23796660 DOI: 10.1016/j.neurobiolaging.2013.05.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 04/10/2013] [Accepted: 05/12/2013] [Indexed: 02/08/2023]
Abstract
Neurogenesis persists in the adult brain as a form of plasticity due to the existence of neural stem cells (NSCs). Alterations in neurogenesis have been found in transgenic Alzheimer's disease (AD) mouse models, but NSC activity and neurogenesis in sporadic AD models remains to be examined. We herein describe a remarkable increase in NSC proliferation in the forebrain of SAMP8, a non-transgenic mouse strain that recapitulates the transition from healthy aging to AD. The increase in proliferation is transient, precedes AD-like symptoms such as amyloid beta 1-42 [Aβ(1-42)] increase or gliosis, and is followed by a steep decline at later stages. Interestingly, in vitro studies indicate that secreted Aβ(1-42) and PI3K signaling may account for the early boost in NSC proliferation. Our results highlight the role of soluble Aβ(1-42) peptide and PI3K in the autocrine regulation of NSCs, and further suggest that over-proliferation of NSCs before the appearance of AD pathology may underlie neurogenic failure during the age-related progression of the disease. These findings have implications for therapeutic approaches based on neurogenesis in AD.
Collapse
|
31
|
Fuster-Matanzo A, Llorens-Martín M, Hernández F, Avila J. Role of neuroinflammation in adult neurogenesis and Alzheimer disease: therapeutic approaches. Mediators Inflamm 2013; 2013:260925. [PMID: 23690659 PMCID: PMC3649701 DOI: 10.1155/2013/260925] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 03/12/2013] [Indexed: 01/22/2023] Open
Abstract
Neuroinflammation, a specialized immune response that takes place in the central nervous system, has been linked to neurodegenerative diseases, and specially, it has been considered as a hallmark of Alzheimer disease, the most common cause of dementia in the elderly nowadays. Furthermore, neuroinflammation has been demonstrated to affect important processes in the brain, such as the formation of new neurons, commonly known as adult neurogenesis. For this, many therapeutic approaches have been developed in order to avoid or mitigate the deleterious effects caused by the chronic activation of the immune response. Considering this, in this paper we revise the relationships between neuroinflammation, Alzheimer disease, and adult neurogenesis, as well as the current therapeutic approaches that have been developed in the field.
Collapse
Affiliation(s)
- Almudena Fuster-Matanzo
- Department of Molecular Neurobiology, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED, ISCIII), 28031 Madrid, Spain
| | - María Llorens-Martín
- Department of Molecular Neurobiology, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED, ISCIII), 28031 Madrid, Spain
| | - Félix Hernández
- Department of Molecular Neurobiology, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED, ISCIII), 28031 Madrid, Spain
| | - Jesús Avila
- Department of Molecular Neurobiology, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED, ISCIII), 28031 Madrid, Spain
| |
Collapse
|
32
|
Omega-3 polyunsaturated Fatty acids enhance neuronal differentiation in cultured rat neural stem cells. Stem Cells Int 2013; 2013:490476. [PMID: 23365582 PMCID: PMC3556893 DOI: 10.1155/2013/490476] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 11/30/2012] [Accepted: 12/03/2012] [Indexed: 01/24/2023] Open
Abstract
Polyunsaturated fatty acids (PUFAs) can induce neurogenesis and recovery from brain diseases. However, the exact mechanisms of the beneficial effects of PUFAs have not been conclusively described. We recently reported that docosahexaenoic acid (DHA) induced neuronal differentiation by decreasing Hes1 expression and increasing p27kip1 expression, which causes cell cycle arrest in neural stem cells (NSCs). In the present study, we examined the effect of eicosapentaenoic acid (EPA) and arachidonic acid (AA) on differentiation, expression of basic helix-loop-helix transcription factors (Hes1, Hes6, and NeuroD), and the cell cycle of cultured NSCs. EPA also increased mRNA levels of Hes1, an inhibitor of neuronal differentiation, Hes6, an inhibitor of Hes1, NeuroD, and Map2 mRNA and Tuj-1-positive cells (a neuronal marker), indicating that EPA induced neuronal differentiation. EPA increased the mRNA levels of p21cip1 and p27kip1, a cyclin-dependent kinase inhibitor, which indicated that EPA induced cell cycle arrest. Treatment with AA decreased Hes1 mRNA but did not affect NeuroD and Map2 mRNA levels. Furthermore, AA did not affect the number of Tuj-1-positive cells or cell cycle progression. These results indicated that EPA could be involved in neuronal differentiation by mechanisms alternative to those of DHA, whereas AA did not affect neuronal differentiation in NSCs.
Collapse
|
33
|
Borre Y, Sir V, de Kivit S, Westphal KG, Olivier B, Oosting RS. Minocycline restores spatial but not fear memory in olfactory bulbectomized rats. Eur J Pharmacol 2012; 697:59-64. [DOI: 10.1016/j.ejphar.2012.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Revised: 08/27/2012] [Accepted: 09/06/2012] [Indexed: 12/22/2022]
|
34
|
Furukawa-Hibi Y, Yun J, Nagai T, Yamada K. Transcriptional suppression of the neuronal PAS domain 4 (Npas4) gene by stress via the binding of agonist-bound glucocorticoid receptor to its promoter. J Neurochem 2012; 123:866-75. [PMID: 23020797 DOI: 10.1111/jnc.12034] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 09/20/2012] [Accepted: 09/24/2012] [Indexed: 01/25/2023]
Abstract
Neuronal PAS domain 4 (NPAS4), a brain-specific helix-loop-helix transcription factor, has recently been shown to regulate the development of GABAergic inhibitory neurons. We previously reported that Npas4 mRNA expression levels were reduced in the hippocampus of mice exposed to social isolation or restraint stress, which was accompanied by impairment of memory, emotional behavior, and hippocampal neurogenesis. Therefore, the reduction of NPAS4 expression may play a role in stress-induced brain dysfunction. In this study, to investigate the transcriptional regulation of Npas4 by stress, we focused on the effect of glucocorticoids (GCs) upon Npas4 transcription. Corticosterone treatment reduced Npas4 expression in the frontal cortex and hippocampus, whereas adrenalectomy caused an increase in expression. GC receptor (GR) antagonist, mifepristone, inhibited the stress-induced reduction of Npas4 expression. Putative negative glucocorticoid response elements (GREs) were found -2000 to -1000 upstream of the Npas4 transcription initiation site. Npas4 promoter activity was increased by mifepristone or by mutation of the negative GRE sequences. A chromatin immunoprecipitation assay revealed that restraint stress increased the binding of GR to Npas4 promoter region in the hippocampus. These results suggest that transcription of Npas4 is down-regulated by stress via the binding of agonist-bound GR to its promoter.
Collapse
Affiliation(s)
- Yoko Furukawa-Hibi
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | |
Collapse
|
35
|
Abstract
INTRODUCTION Over the last 8 years, emerging studies bridging the gap between nutrition and mental health have resolutely established that learning and memory abilities as well as mood can be influenced by diet. However, the mechanisms by which diet modulates mental health are still not well understood. Sources of data In this article, a review of the literature was conducted using PubMed to identify studies that provide functional implications of adult hippocampal neurogenesis (AHN) and its modulation by diet. AREAS OF AGREEMENT One of the brain structures associated with learning and memory as well as mood is the hippocampus. Importantly, the hippocampus is one of the two structures in the adult brain where the formation of newborn neurons, or neurogenesis, persists. AREAS OF CONTROVERSY The exact roles of these newborn neurons in learning, memory formation and mood regulation remain elusive. GROWING POINTS Nevertheless, there has been accumulating evidence linking cognition and mood to neurogenesis occurring in the adult hippocampus. Therefore, modulation of AHN by diet emerges as a possible mechanism by which nutrition impacts on mental health. AREAS TIMELY FOR DEVELOPING RESEARCH This area of investigation is new and needs attention because a better understanding of the neurological mechanisms by which nutrition affect mental health may lead to novel dietary approaches for disease prevention, healthier ageing and discovery of new therapeutic targets for mental illnesses.
Collapse
|
36
|
Tang SW, Helmeste D, Leonard B. Is neurogenesis relevant in depression and in the mechanism of antidepressant drug action? A critical review. World J Biol Psychiatry 2012; 13:402-12. [PMID: 22175526 DOI: 10.3109/15622975.2011.639800] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Major depression is a complex disorder that involves genetic, epigenetic and environmental factors in its aetiology. Recent research has suggested that hippocampal neurogenesis may play a role in antidepressant action. However, careful examination of the literature suggests that the complex biological and psychological changes associated with depression cannot be attributed to disturbance in hippocampal neurogenesis alone. While antidepressants may induce hippocampal neurogenesis in non-human primates, there is a paucity of evidence that such effects are sufficient for full therapeutic action in humans. METHODS This review examines the literature on neurogenesis and discusses the stress-induced cortisol neurotoxicity and antidepressant-induced neurogenesis rescue model of depression. The disparity between a simple antidepressant-induced neurogenesis rescue model in the hippocampus and the complexity of clinical depression is analyzed through critical evaluation of recent research data. RESULTS AND CONCLUSIONS Major depression is a complex brain disorder with multiple symptoms and disturbances reflecting dysfunction in more than one single brain area. Initial research suggesting a model of hippocampal degeneration as basis of depression, and reversal by antidepressants through neurogenesis seems to be over-simplified given the emergence of new data. Synaptogenesis and re-organization or re-integration of new neurons rather than simple addition of new neurons may underlie the role of antidepressant drugs in the reversal of some but not all symptoms in depression. The importance of the neurogenesis hypothesis of depression and antidepressant action lies in stimulating further research into the possible roles played by the new neurons and synapses generated.
Collapse
Affiliation(s)
- Siu W Tang
- Department of Psychiatry, University of California, Irvine, CA 92697-1681, USA.
| | | | | |
Collapse
|
37
|
Sinha M, Mukhopadhyay S, Bhattacharyya NP. Mechanism(s) of alteration of micro RNA expressions in Huntington's disease and their possible contributions to the observed cellular and molecular dysfunctions in the disease. Neuromolecular Med 2012; 14:221-43. [PMID: 22581158 DOI: 10.1007/s12017-012-8183-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 04/24/2012] [Indexed: 12/27/2022]
Abstract
To identify the mechanism of deregulation of micro RNAs (miRNAs) altered in Huntington's disease (HD) and their possible contributions to the altered cellular and molecular functions observed in the disease, we analyzed the altered miRNAs in the postmortem brains of HD patients. There are 54 miRNAs differentially expressed in HD brains of which 30 are upregulated and 24 downregulated. Some of these miRNAs were also altered in various models of the disease. Regulation of these miRNAs was attributed to transcription factors and the host genes to which these miRNAs reside. We observed that transcription regulators TP53, E2F1, REST, and GATA4 together could regulate expressions of 26 miRNAs in HD. Altered expressions of 13 intronic miRNAs were correlated with the expressions of their host genes. From literature, we further collected 287 experimentally validated targets of miRNAs upregulated in HD, while 304 validated targets of downregulated miRNAs in HD. Analysis of these validated target genes of altered miRNAs by gene ontology (GO) revealed that these genes are significantly enriched in GO terms belonging to (1) apoptosis, (2) differentiation and development, (3) fatty acid, cholesterol, lipid, glucose, and carbohydrate metabolism, (4) cell cycle and growth, and (5) transcription regulation. Experimental evidences that these processes are altered in HD are provided from published reports. In conclusion, altered miRNAs in HD might target many genes and may contribute to the altered cellular and molecular functions observed in HD.
Collapse
Affiliation(s)
- Mithun Sinha
- Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhan Nagar, Kolkata, 700 064, India
| | | | | |
Collapse
|
38
|
Swayne LA, Wicki-Stordeur L. Ion channels in postnatal neurogenesis: potential targets for brain repair. Channels (Austin) 2012; 6:69-74. [PMID: 22614818 DOI: 10.4161/chan.19721] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Neural stem and progenitor cells (NSC/NPCs) are unspecialized cells found in the adult peri-ventricular and sub-granular zones that are capable of self-renewal, migration, and differentiation into new neurons through the remarkable process of postnatal neurogenesis. We are now beginning to understand that the concerted action of ion channels, multi-pass transmembrane proteins that allow passage of ions across otherwise impermeable cellular membranes tightly regulate this process. Specific ion channels control proliferation, differentiation and survival. Furthermore, they have the potential to be highly selective drug targets due to their complex structures. As such, these proteins represent intriguing prospects for control and optimization of postnatal neurogenesis for neural regeneration following brain injury or disease. Here, we concentrate on ion channels identified in adult ventricular zone NSC/NPCs that have been found to influence the stages of neurogenesis. Finally, we outline the potential of these channels to elicit repair, and highlight the outstanding challenges.
Collapse
Affiliation(s)
- Leigh Anne Swayne
- Division of Medical Sciences; Island Medical Program, University of Victoria, Victoria, BC, Canada.
| | | |
Collapse
|
39
|
Borre Y, Bosman E, Lemstra S, Westphal KG, Olivier B, Oosting RS. Memantine partly rescues behavioral and cognitive deficits in an animal model of neurodegeneration. Neuropharmacology 2012; 62:2010-7. [PMID: 22248638 DOI: 10.1016/j.neuropharm.2011.12.034] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 12/05/2011] [Accepted: 12/26/2011] [Indexed: 12/21/2022]
Abstract
Memantine, a non-competitive NMDA receptor antagonist, is used for the treatment of Alzheimer's disease (AD) and off-label as an anti-depressant. Here we investigated possible anti-depressant, cognitive enhancing and neuroprotective effects of memantine in the olfactory bulbectomized (OBX) rat. OBX is used as a screening model for antidepressants and shows cognitive disturbances. In Experiment I, memantine treatment started 14 days after OBX surgery (this setup is similar to what we use for screening of potential antidepressants) and 2 days before surgery in experiment II. In both experiments, memantine (20 mg/kg, p.o) was administered once daily for 28 days. Animals were tested in the open field (locomotor activity), passive avoidance (fear learning and memory), and holeboard (spatial acquisition and memory) before and after the bulbectomy. Memantine, when administered before surgery, prevented OBX-induced hyperactivity and partly fear memory loss. These behavioral effects were present for at least 3 weeks after cessation of treatment. Memantine, however did not improve spatial memory. When administered 2 weeks after OBX surgery, memantine was ineffective in normalizing open field hyperactivity and improving cognitive deficits. Interestingly, after the animals were retrained in passive avoidance, memantine- treated OBX rats (both in experiment I and II) showed improved fear learning and memory. Our findings suggest that memantine has both neuroprotective and cognitive enhancing effects without antidepressant-like properties in the OBX rat. Based on our results, we propose that memantine may be more beneficial to AD patients when administered early in the disease process.
Collapse
Affiliation(s)
- Yuliya Borre
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences and Rudolf Magnus Institute of Neuroscience, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands.
| | | | | | | | | | | |
Collapse
|
40
|
Papanikolopoulou K, Skoulakis EMC. The power and richness of modelling tauopathies in Drosophila. Mol Neurobiol 2011; 44:122-33. [PMID: 21681411 DOI: 10.1007/s12035-011-8193-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 06/06/2011] [Indexed: 02/07/2023]
Abstract
Tauopathies are a group of neurodegenerative disorders characterised by altered levels of phosphorylation or mutations in the neuronal microtubule protein Tau. The heterogeneous pathology of tauopathies suggests differential susceptibility of different neuronal types to wild-type and mutant Tau. The genetic power and facility of the Drosophila model has been instrumental in exploring the molecular aetiologies of tauopathies, identifying additional proteins likely contributing to neuronal dysfunction and toxicity and novel Tau phosphorylations mediating them. Importantly, recent results indicate tissue- and temporal-specific effects on dysfunction and toxicity coupled with differential effects of distinct Tau isoforms within them. Therefore, they reveal an unexpected richness of the Drosophila model that, coupled with its molecular genetic power, will likely play a significant role in our understanding of multiple tauopathies potentially leading to their differential treatment.
Collapse
Affiliation(s)
- Katerina Papanikolopoulou
- Institute of Cellular and Developmental Biology, Biomedical Sciences Research Centre Alexander Fleming, Vari 16672, Greece.
| | | |
Collapse
|
41
|
Galan L, Gomez-Pinedo U, Vela-Souto A, Guerrero-Sola A, Barcia JA, Gutierrez AR, Martinez-Martinez A, Jiménez MSB, García-Verdugo JM, Matias-Guiu J. Subventricular zone in motor neuron disease with frontotemporal dementia. Neurosci Lett 2011; 499:9-13. [PMID: 21616125 DOI: 10.1016/j.neulet.2011.05.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Revised: 04/06/2011] [Accepted: 05/09/2011] [Indexed: 10/18/2022]
Abstract
Investigate how the subventricular proliferation and organisation is modified in a patient with FTLD-ALS. We studied the subventricular zone (SVZ) of a patient with FTLD-ALS immunohistochemical and histologically. We found an increase of Ki-67 positive cells and neuroblast in the subventricular zone, suggesting an activation of proliferating activity in response to FTD-ALS. This proliferation can act as a compensatory mechanism for rapid neuronal death and its modulation could provide a new therapeutic pathway in ALS. These results suggest a modification of neurogenesis in FTD-ALS.
Collapse
Affiliation(s)
- Lucia Galan
- Neuromuscular Unit, Neurology Department, Hospital Clinico San Carlos, Madrid, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Zunszain PA, Anacker C, Cattaneo A, Carvalho LA, Pariante CM. Glucocorticoids, cytokines and brain abnormalities in depression. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:722-9. [PMID: 20406665 PMCID: PMC3513408 DOI: 10.1016/j.pnpbp.2010.04.011] [Citation(s) in RCA: 361] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Revised: 04/07/2010] [Accepted: 04/14/2010] [Indexed: 02/09/2023]
Abstract
Major depression (MD) is a common psychiatric disorder with a complex and multifactor aetiology. Potential mechanisms associated with the pathogenesis of this disorder include monoamine deficits, hypothalamic-pituitary-adrenal (HPA) axis dysfunctions, inflammatory and/or neurodegenerative alterations. An increased secretion and reactivity of cortisol together with an altered feedback inhibition are the most widely observed HPA abnormalities in MD patients. Glucocorticoids, such as cortisol, are vital hormones that are released in response to stress, and regulate metabolism and immunity but also neuronal survival and neurogenesis. Interestingly depression is highly prevalent in infectious, autoimmune and neurodegenerative diseases and at the same time, depressed patients show higher levels of pro-inflammatory cytokines. Since communication occurs between the endocrine, immune and central nervous system, an activation of the inflammatory responses can affect neuroendocrine processes, and vice versa. Therefore, HPA axis hyperactivity and inflammation might be part of the same pathophysiological process: HPA axis hyperactivity is a marker of glucocorticoid resistance, implying ineffective action of glucocorticoid hormones on target tissues, which could lead to immune activation; and, equally, inflammation could stimulate HPA axis activity via both a direct action of cytokines on the brain and by inducing glucocorticoid resistance. In addition, increased levels of pro-inflammatory cytokines also induce the production of neurotoxic end products of the tryptophan-kynurenine pathway. Although the evidence for neurodegeneration in MD is controversial, depression is co-morbid with many other conditions where neurodegeneration is present. Since several systems seem to be involved interacting with each other, we cannot unequivocally accept the simple model that glucocorticoids induce neurodegeneration, but rather that elevated cytokines, in the context of glucocorticoid resistance, are probably the offenders. Chronic inflammatory changes in the presence of glucocorticoid resistance may represent a common feature that could be responsible for the enhanced vulnerability of depressed patients to develop neurodegenerative changes later in life. However, further studies are needed to clarify the relative contribution of glucocorticoids and inflammatory signals to MD and other disorders.
Collapse
Affiliation(s)
- Patricia A. Zunszain
- Stress, Psychiatry and Immunology Laboratory (SPI-Lab), Department of Psychological Medicine, Institute of Psychiatry, King’s College London, 125 Coldharbour Lane, SE5 9NU, London, United Kingdom
,Corresponding author. (P.A. Zunszain).
| | - Christoph Anacker
- Stress, Psychiatry and Immunology Laboratory (SPI-Lab), Department of Psychological Medicine, Institute of Psychiatry, King’s College London, 125 Coldharbour Lane, SE5 9NU, London, United Kingdom
| | - Annamaria Cattaneo
- Stress, Psychiatry and Immunology Laboratory (SPI-Lab), Department of Psychological Medicine, Institute of Psychiatry, King’s College London, 125 Coldharbour Lane, SE5 9NU, London, United Kingdom
,Division of Biology and Genetics, Department of Biomedical Sciences and Biotechnology, University of Brescia, Brescia, Italy
| | - Livia A. Carvalho
- Stress, Psychiatry and Immunology Laboratory (SPI-Lab), Department of Psychological Medicine, Institute of Psychiatry, King’s College London, 125 Coldharbour Lane, SE5 9NU, London, United Kingdom
| | - Carmine M. Pariante
- Stress, Psychiatry and Immunology Laboratory (SPI-Lab), Department of Psychological Medicine, Institute of Psychiatry, King’s College London, 125 Coldharbour Lane, SE5 9NU, London, United Kingdom
| |
Collapse
|
43
|
Lemmens MAM, Sierksma ASR, Rutten BPF, Dennissen F, Steinbusch HWM, Lucassen PJ, Schmitz C. Age-related changes of neuron numbers in the frontal cortex of a transgenic mouse model of Alzheimer's disease. Brain Struct Funct 2011; 216:227-37. [PMID: 21409417 PMCID: PMC3155024 DOI: 10.1007/s00429-011-0305-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Accepted: 02/22/2011] [Indexed: 12/16/2022]
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder, characterized by amyloid plaque accumulation, intracellular tangles and neuronal loss in selective brain regions. The frontal cortex, important for executive functioning, is one of the regions that are affected. Here, we investigated the neurodegenerative effects of mutant human amyloid precursor protein (APP) and presenilin 1 (PS1) on frontal cortex neurons in APP/PS1KI mice, a transgenic mouse model of AD, expressing two mutations in the human APP, as well as two human PS1 mutations knocked-in into the mouse PS1 gene in a homozygous (ho) manner. Although the hippocampus is significantly affected in these mice, very little is known about the effects of these mutations on selective neuronal populations and plaque load in the frontal cortex. In this study, cytoarchitectural changes were characterized using high precision design-based stereology to evaluate plaque load, total neuron numbers, as well as total numbers of parvalbumin- (PV) and calretinin- (CR) immunoreactive (ir) neurons in the frontal cortex of 2- and 10-month-old APP/PS1KI mice. The frontal cortex was divided into two subfields: layers II–IV and layers V–VI, the latter of which showed substantially more extracellular amyloid-beta aggregates. We found a 34% neuron loss in layers V–VI in the frontal cortex of 10-month-old APP/PS1KI mice compared to 2-month-old, while there was no change in PV- and CR-ir neurons in these mice. In addition, the plaque load in layers V–VI of 10-month-old APP/PS1KI mice was only 11% and did not fully account for the extent of neuronal loss. Interestingly, an increase was found in the total number of PV-ir neurons in all frontal cortical layers of single transgenic APP mice and in layers II–IV of single transgenic PS1ho mice between 2 and 10 months of age. In conclusion, the APP/PS1KI mice provide novel insights into the regional selective vulnerability in the frontal cortex during AD that, together with previous findings in the hippocampus, are remarkably similar to the human situation.
Collapse
Affiliation(s)
- Marijke A M Lemmens
- Department for Neuroscience, Faculty of Health, Medicine and Life Sciences (FHML), School for Mental Health and Neuroscience (MHeNS), European Graduate School of Neuroscience (EURON), Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
44
|
Chen X, Tolkovsky AM, Herbert J. Cell origin and culture history determine successful integration of neural precursor transplants into the dentate gyrus of the adult rat. PLoS One 2011; 6:e17072. [PMID: 21359219 PMCID: PMC3040198 DOI: 10.1371/journal.pone.0017072] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Accepted: 01/17/2011] [Indexed: 11/18/2022] Open
Abstract
The success of transplants of neural tissue into the adult dentate gyrus in generating mature neurons is highly variable. Here we address the roles of the origin of the tissue and its pre-implantation preparation, and show that both are critical. We transplanted neonatal cultured or primary rat cells from either the ventral subventricular zone (vSVZ) or the dentate gyrus (DG) into the adult rat DG. Only primary DG cells robustly generated DG neurons (80% NeuN and Prox1-positive cells at 6 weeks), substantially repaired the damaged DG, and formed glutamatergic projections to the target CA3 region. Cultured DG cells expanded for 7 days showed limited neuronal differentiation after transplantation (10% NeuN and Prox1-positive cells) whereas cultured or primary vSVZ cells failed to make any Prox1-positive DG granular neurons. We found that a specific population of postmitotic young neurons (triple doublecortin/NeuN/Prox1-positive) were particularly abundant in primary DG cells, but were markedly reduced in the cultured DG cells and were absent in the cultured and primary vSVZ cells. Labelling of primary DG cells with the mitotic marker BrdU suggested that postmitotic young neurons are the source of the transplanted mature neurons in-vivo. We conclude that both the origin and pre-transplantation history of donor cells are key factors that determine the outcome of transplantation. These findings may be of therapeutic interest for cell replacement therapy in treating the damaged hippocampus.
Collapse
Affiliation(s)
- Xia Chen
- Cambridge Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | - Aviva M. Tolkovsky
- Cambridge Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | - Joe Herbert
- Cambridge Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
45
|
Cholinergic influences on cortical development and adult neurogenesis. Behav Brain Res 2011; 221:379-88. [PMID: 21272598 DOI: 10.1016/j.bbr.2011.01.021] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Revised: 01/10/2011] [Accepted: 01/13/2011] [Indexed: 11/22/2022]
Abstract
In this review, we focus on immature neurons and their regulation by the cholinergic system, both during cortical development as well as during adult neurogenesis. We discuss various studies that indicate roles for acetylcholine in precursor development and neuronal differentiation. Cholinergic neurons projecting from the basal forebrain innervate the cerebral cortex during critical periods of neuronal development. Acetylcholine stimulation may help to promote a favourable environment for neuronal maturation. Afferents and their cortical target cells interact and are likely to influence each other during the establishment and refinement of connections. Intracortical cholinergic interneurons similarly have a local effect on cortical circuits. Reduced cholinergic innervation during development hence leads to reduced cortical thickness and dendritic abnormalities. Acetylcholine is also likely to play a critical role in neuronal plasticity, as shown in the visual and barrel cortices. Spontaneous nicotinic excitation is also important during a brief developmental window in the first postnatal weeks leading to waves of neural activity, likely to have an effect on neurite extension, target selection and synaptogenesis. In the hippocampus such activity plays a role in the maturation of GABAergic synapses during the developmental shift from depolarizing to hyperpolarizing transmission. The cholinergic system also seems likely to regulate hippocampal neurogenesis in the adult, positively promoting proliferation, differentiation, integration and potentially survival of newborn neurons.
Collapse
|
46
|
Phosphorylation differentiates tau-dependent neuronal toxicity and dysfunction. Biochem Soc Trans 2010; 38:981-7. [PMID: 20658989 DOI: 10.1042/bst0380981] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The heterogeneous pathology of tauopathies and the differential susceptibility of different neuronal types to WT (wild-type) and mutant tau suggest that phosphorylation at particular sites rather than hyperphosphorylation mediates toxicity or dysfunction in a cell-type-specific manner. Pan-neuronal accumulation of tau in the Drosophila CNS (central nervous system) specifically affected the MBs (mushroom body neurons), consistent with neuronal type-specific effects. The MB aberrations depended, at least in part, on occupation of two novel phosphorylation sites: Ser(238) and Thr(245). The degree of isoform-specific MB aberrations was paralleled by defects in associative learning, as blocking putative Ser(238) and Thr(245) phosphorylation yielded structurally normal, but profoundly dysfunctional, MBs, as animals accumulating the mutant protein exhibited strongly impaired associative learning. Similarly dysfunctional MBs were obtained by temporally restricting tau accumulation to the adult CNS, which also altered the tau phosphorylation pattern. Our data clearly distinguish tau-dependent neuronal degeneration and dysfunction and suggest that temporal differences in occupation of the same phosphorylation sites are likely to mediate these distinct effects of tau.
Collapse
|
47
|
van Tijn P, Kamphuis W, Marlatt MW, Hol EM, Lucassen PJ. Presenilin mouse and zebrafish models for dementia: focus on neurogenesis. Prog Neurobiol 2010; 93:149-64. [PMID: 21056616 DOI: 10.1016/j.pneurobio.2010.10.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 10/27/2010] [Accepted: 10/31/2010] [Indexed: 01/18/2023]
Abstract
Autosomal dominant mutations in the presenilin gene PSEN cause familial Alzheimer's disease (AD), a neurological disorder pathologically characterized by intraneuronal accumulation and extracellular deposition of amyloid-β in plaques and intraneuronal, hyperphosphorylated tau aggregation in neurofibrillary tangles. Presenilins (PS/PSENs) are part of the proteolytic γ-secretase complex, which cleaves substrate proteins within the membrane. Cleavage of the amyloid precursor protein (APP) by γ-secretase releases amyloid-β peptides. Besides its role in the processing of APP and other transmembrane proteins, presenilin plays an important role in neural progenitor cell maintenance and neurogenesis. In this review, we discuss the role of presenilin in relation to neurogenesis and neurodegeneration and review the currently available presenilin animal models. In addition to established mouse models, zebrafish are emerging as an attractive vertebrate model organism to study the role of presenilin during the development of the nervous system and in neurodegenerative disorders involving presenilin. Zebrafish is a suitable model organism for large-scale drug screening, making this a valuable model to identify novel therapeutic targets for AD.
Collapse
Affiliation(s)
- Paula van Tijn
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
48
|
Swayne LA, Sorbara CD, Bennett SAL. Pannexin 2 is expressed by postnatal hippocampal neural progenitors and modulates neuronal commitment. J Biol Chem 2010; 285:24977-86. [PMID: 20529862 DOI: 10.1074/jbc.m110.130054] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The pannexins (Panx1, -2, and -3) are a mammalian family of putative single membrane channels discovered through homology to invertebrate gap junction-forming proteins, the innexins. Because connexin gap junction proteins are known regulators of neural stem and progenitor cell proliferation, migration, and specification, we asked whether pannexins, specifically Panx2, play a similar role in the postnatal hippocampus. We show that Panx2 protein is differentially expressed by multipotential progenitor cells and mature neurons. Both in vivo and in vitro, Type I and IIa stem-like neural progenitor cells express an S-palmitoylated Panx2 species localizing to Golgi and endoplasmic reticulum membranes. Protein expression is down-regulated during neurogenesis in neuronally committed Type IIb and III progenitor cells and immature neurons. Panx2 is re-expressed by neurons following maturation. Protein expressed by mature neurons is not palmitoylated and localizes to the plasma membrane. To assess the impact of Panx2 on neuronal differentiation, we used short hairpin RNA to suppress Panx2 expression in Neuro2a cells. Knockdown significantly accelerated the rate of neuronal differentiation. Neuritic extension and the expression of antigenic markers of mature neurons occurred earlier in stable lines expressing Panx2 short hairpin RNA than in controls. Together, these findings describe an endogenous post-translational regulation of Panx2, specific to early neural progenitor cells, and demonstrate that this expression plays a role in modulating the timing of their commitment to a neuronal lineage.
Collapse
Affiliation(s)
- Leigh Anne Swayne
- Department of Biochemistry, Neural Regeneration Laboratory and Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario K1M 1E5, Canada.
| | | | | |
Collapse
|
49
|
Differential effects of Tau on the integrity and function of neurons essential for learning in Drosophila. J Neurosci 2010; 30:464-77. [PMID: 20071510 DOI: 10.1523/jneurosci.1490-09.2010] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Tauopathies are a heterogeneous group of neurodegenerative dementias involving perturbations in the levels, phosphorylation, or mutations of the microtubule-binding protein Tau. The heterogeneous pathology in humans and model organisms suggests differential susceptibility of neuronal types to wild-type (WT) and mutant Tau. WT and mutant human Tau-encoding transgenes expressed pan-neuronally in the Drosophila CNS yielded specific and differential toxicity in the embryonic neuroblasts that generate the mushroom body (MB) neurons, suggesting cell type-specific effects of Tau in the CNS. Frontotemporal dementia with parkinsonism-17-linked mutant isoforms were significantly less toxic in MB development. Tau hyperphosphorylation was essential for these MB aberrations, and we identified two novel putative phosphorylation sites, Ser(238) and Thr(245), on WT hTau essential for its toxic effects on MB integrity. Significantly, blocking putative Ser(238) and Thr(245) phosphorylation yielded animals with apparently structurally normal but profoundly dysfunctional MBs, because animals accumulating this mutant protein exhibited strongly impaired associative learning. Interestingly, the mutant protein was hyperphosphorylated at epitopes typically associated with toxicity and neurodegeneration, such as AT8, AT100, and the Par-1 targets Ser(262) and Ser(356), suggesting that these sites in the context of adult intact MBs mediate dysfunction and occupation of these sites may precede the toxicity-associated Ser(238) and Thr(245) phosphorylation. The data support the notion that phosphorylation at particular sites rather than hyperphosphorylation per se mediates toxicity or dysfunction in a cell type-specific manner.
Collapse
|
50
|
DeCarolis NA, Eisch AJ. Hippocampal neurogenesis as a target for the treatment of mental illness: a critical evaluation. Neuropharmacology 2010; 58:884-93. [PMID: 20060007 DOI: 10.1016/j.neuropharm.2009.12.013] [Citation(s) in RCA: 191] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Revised: 12/09/2009] [Accepted: 12/14/2009] [Indexed: 12/21/2022]
Abstract
Over one-quarter of adult Americans are diagnosed with a mental illness like Major Depressive Disorder (MDD), Post-Traumatic Stress Disorder (PTSD), schizophrenia, and Alzheimer's Disease. In addition to the exceptional personal burden these disorders exert on patients and their families, they also have enormous cost to society. Although existing pharmacological and psychosocial treatments alleviate symptoms in many patients, the comorbidity, severity, and intractable nature of mental disorders strongly underscore the need for novel strategies. As the hippocampus is a site of structural and functional pathology in most mental illnesses, a hippocampal-based treatment approach has been proposed to counteract the cognitive deficits and mood dysregulation that are hallmarks of psychiatric disorders. In particular, preclinical and clinical research suggests that hippocampal neurogenesis, the generation of new neurons in the adult dentate gyrus, may be harnessed to treat mental illness. There are obvious applications and allures of this approach; for example, perhaps stimulating hippocampal neurogenesis would reverse the overt and noncontroversial hippocampal atrophy and functional deficits observed in Alzheimer's Disease and schizophrenia, or the more controversial hippocampal deficits seen in MDD and PTSD. However, critical examination suggests that neurogenesis may only correlate with mental illness and treatment, suggesting targeting neurogenesis alone is not a sufficient treatment strategy. Here we review the classic and causative links between adult hippocampal neurogenesis and mental disorders, and provide a critical evaluation of how (and if) our basic knowledge of new neurons in the adult hippocampus might eventually help combat or even prevent mental illness.
Collapse
Affiliation(s)
- Nathan A DeCarolis
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX 75390-9070, USA
| | | |
Collapse
|