1
|
Adamczyk PM, Shaw A, Morella IM, More L. Neurobiology, molecular pathways, and environmental influences in antisocial traits and personality disorders. Neuropharmacology 2025; 269:110322. [PMID: 39864585 DOI: 10.1016/j.neuropharm.2025.110322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 12/17/2024] [Accepted: 01/20/2025] [Indexed: 01/28/2025]
Abstract
Personality disorders (PDs) are psychiatric conditions characterized by enduring patterns of cognition, emotion, and behaviour that deviate significantly from cultural norms, causing distress or impairment. The aetiology of PDs is complex, involving both genetic and environmental factors. Genetic studies estimate the heritability of PDs at 30%-60%, implicating genes involved in neurotransmitter regulation, such as those for serotonin transporters and dopamine receptors. Environmental factors, including childhood trauma and chronic stress, interact with genetic predispositions to induce epigenetic modifications like DNA methylation and histone modifications, contributing to PD development. Neurobiological research has identified structural and functional abnormalities in brain regions related to emotional regulation and social cognition, such as the amygdala, prefrontal cortex, and limbic system. These abnormalities are linked to impaired emotion processing and interpersonal functioning in PDs. This review focuses on how environmental factors shape maladaptive behaviours and endophenotypes central to many PDs. It explores the interaction between the Ras-ERK, p38, and mTOR molecular pathways in response to environmental stimuli, and examines the role of oxidative stress and mitochondrial metabolism in these processes. Also reviewed are various types of PDs and existing animal models that replicate key endophenotypes, highlighting changes in neurotransmitters and neurohormones. Identifying molecular biomarkers can lead to the development of "enviromimetic" drugs, which mimic environmental influences to activate molecular pathways, facilitating targeted, personalized treatments based on the molecular profiles of individuals with PDs. Ultimately, understanding the molecular mechanisms of PDs promises to enhance diagnostic accuracy, prognosis, and therapeutic outcomes for affected individuals.
Collapse
Affiliation(s)
- Patryk M Adamczyk
- School of Pharmacy and Biomedical Sciences, The University of Central Lancashire, Preston, UK
| | - Andrew Shaw
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, UK.
| | - Ilaria M Morella
- University of Pavia, Department of Biology and Biotechnology "Lazzaro Spallanzani", Pavia, Italy; Cardiff University, School of Medicine, Division of Psychological Medicine and Clinical Neurosciences, Cardiff, UK.
| | - Lorenzo More
- School of Pharmacy and Biomedical Sciences, The University of Central Lancashire, Preston, UK.
| |
Collapse
|
2
|
Morè L, Privitera L, Lopes M, Arthur JSC, Lauterborn JC, Corrêa SAL, Frenguelli BG. MSK1 is required for the experience- and ampakine-dependent enhancement of spatial reference memory and reversal learning and for the induction of Arc and BDNF. Neuropharmacology 2024; 261:110110. [PMID: 39128584 DOI: 10.1016/j.neuropharm.2024.110110] [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: 02/27/2024] [Revised: 08/08/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
There is considerable interest in the development of nootropics, pharmacological agents that can improve cognition across a range of both cognitive modalities and cognitive disabilities. One class of cognitive enhancers, the ampakines, has attracted particular attention by virtue of improving cognition associated with animal models of neurodevelopmental, neurodegenerative, and psychiatric conditions, as well as in age-related cognitive impairment. Ampakines elevate CNS levels of BDNF, and it is through this elevation that their beneficial actions are believed to occur. However, what transduces the elevation of BDNF into long-lasting cognitive enhancement is not known. We have previously shown that MSK1, by virtue of its ability to regulate gene transcription, converts the elevation of BDNF associated with environmental enrichment into molecular, synaptic, cognitive and genomic adaptations that underlie enrichment-induced enhanced synaptic plasticity and learning and memory, a property that MSK1 retains across the lifespan. To establish whether MSK1 similarly converts ampakine-induced elevations of BDNF into cognitive enhancement we tested an ampakine (CX929) in male WT mice and in male mice in which the kinase activity of MSK1 was inactivated. We found that MSK1 is required for the ampakine-dependent improvement in spatial reference memory and cognitive flexibility, and for the elevations of BDNF and the plasticity-related protein Arc associated with ampakines and experience. These observations implicate MSK1 as a key enabler of the beneficial effects of ampakines on cognitive function, and furthermore identify MSK1 as a hub for BDNF-elevating nootropic strategies.
Collapse
Affiliation(s)
- Lorenzo Morè
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK; School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, PR1 2HE, UK
| | - Lucia Privitera
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Marcia Lopes
- Bradford School of Pharmacy and Medical Sciences, University of Bradford, Bradford, BD7 1DP, UK
| | - J Simon C Arthur
- Division of Cell Signalling and Immunology, University of Dundee, Dundee, DD1 5EH, UK
| | - Julie C Lauterborn
- Department of Anatomy & Neurobiology, University of California, Irvine, USA
| | - Sonia A L Corrêa
- Bradford School of Pharmacy and Medical Sciences, University of Bradford, Bradford, BD7 1DP, UK; Department of Life Sciences, Manchester Metropolitan University, Manchester, M15 6BH, UK
| | | |
Collapse
|
3
|
Bommaraju S, Dhokne MD, Arun EV, Srinivasan K, Sharma SS, Datusalia AK. An insight into crosstalk among multiple signalling pathways contributing to the pathophysiology of PTSD and depressive disorders. Prog Neuropsychopharmacol Biol Psychiatry 2024; 131:110943. [PMID: 38228244 DOI: 10.1016/j.pnpbp.2024.110943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/18/2024]
Abstract
Post-traumatic stress disorder (PTSD) and depressive disorders represent two significant mental health challenges with substantial global prevalence. These are debilitating conditions characterized by persistent, often comorbid, symptoms that severely impact an individual's quality of life. Both PTSD and depressive disorders are often precipitated by exposure to traumatic events or chronic stress. The profound impact of PTSD and depressive disorders on individuals and society necessitates a comprehensive exploration of their shared and distinct pathophysiological features. Although the activation of the stress system is essential for maintaining homeostasis, the ability to recover from it after diminishing the threat stimulus is also equally important. However, little is known about the main reasons for individuals' differential susceptibility to external stressful stimuli. The solution to this question can be found by delving into the interplay of stress with the cognitive and emotional processing of traumatic incidents at the molecular level. Evidence suggests that dysregulation in these signalling cascades may contribute to the persistence and severity of PTSD and depressive symptoms. The treatment strategies available for this disorder are antidepressants, which have shown good efficiency in normalizing symptom severity; however, their efficacy is limited in most individuals. This calls for the exploration and development of innovative medications to address the treatment of PTSD. This review delves into the intricate crosstalk among multiple signalling pathways implicated in the development and manifestation of these mental health conditions. By unravelling the complexities of crosstalk among multiple signalling pathways, this review aims to contribute to the broader knowledge base, providing insights that could inform the development of targeted interventions for individuals grappling with the challenges of PTSD and depressive disorders.
Collapse
Affiliation(s)
- Sumadhura Bommaraju
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Uttar Pradesh (UP) 226002, India
| | - Mrunali D Dhokne
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Uttar Pradesh (UP) 226002, India
| | - E V Arun
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Uttar Pradesh (UP) 226002, India
| | - Krishnamoorthy Srinivasan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), SAS Nagar, Punjab 160062, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), SAS Nagar, Punjab 160062, India
| | - Ashok Kumar Datusalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Uttar Pradesh (UP) 226002, India; Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Raebareli, Uttar Pradesh (UP) 226002, India.
| |
Collapse
|
4
|
Morè L, Privitera L, Cooper DD, Tsogka M, Arthur JSC, Frenguelli BG. MSK1 is required for the beneficial synaptic and cognitive effects of enriched experience across the lifespan. Aging (Albany NY) 2023; 15:6031-6072. [PMID: 37432063 PMCID: PMC10373962 DOI: 10.18632/aging.204833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/31/2023] [Indexed: 07/12/2023]
Abstract
Positive experiences, such as social interaction, cognitive training and physical exercise, have been shown to ameliorate some of the harms to cognition associated with ageing. Animal models of positive interventions, commonly known as environmental enrichment, strongly influence neuronal morphology and synaptic function and enhance cognitive performance. While the profound structural and functional benefits of enrichment have been appreciated for decades, little is known as to how the environment influences neurons to respond and adapt to these positive sensory experiences. We show that adult and aged male wild-type mice that underwent a 10-week environmental enrichment protocol demonstrated improved performance in a variety of behavioural tasks, including those testing spatial working and spatial reference memory, and an enhancement in hippocampal LTP. Aged animals in particular benefitted from enrichment, performing spatial memory tasks at levels similar to healthy adult mice. Many of these benefits, including in gene expression, were absent in mice with a mutation in an enzyme, MSK1, which is activated by BDNF, a growth factor implicated in rodent and human cognition. We conclude that enrichment is beneficial across the lifespan and that MSK1 is required for the full extent of these experience-induced improvements of cognitive abilities, synaptic plasticity and gene expression.
Collapse
Affiliation(s)
- Lorenzo Morè
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK
| | - Lucia Privitera
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Daniel D. Cooper
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Marianthi Tsogka
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| | | | | |
Collapse
|
5
|
The Molecular Effects of Environmental Enrichment on Alzheimer's Disease. Mol Neurobiol 2022; 59:7095-7118. [PMID: 36083518 PMCID: PMC9616781 DOI: 10.1007/s12035-022-03016-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/23/2022] [Indexed: 12/02/2022]
Abstract
Environmental enrichment (EE) is an environmental paradigm encompassing sensory, cognitive, and physical stimulation at a heightened level. Previous studies have reported the beneficial effects of EE in the brain, particularly in the hippocampus. EE improves cognitive function as well as ameliorates depressive and anxiety-like behaviors, making it a potentially effective neuroprotective strategy against neurodegenerative diseases such as Alzheimer's disease (AD). Here, we summarize the current evidence for EE as a neuroprotective strategy as well as the potential molecular pathways that can explain the effects of EE from a biochemical perspective using animal models. The effectiveness of EE in enhancing brain activity against neurodegeneration is explored with a view to differences present in early and late life EE exposure, with its potential application in human being discussed. We discuss EE as one of the non pharmacological approaches in preventing or delaying the onset of AD for future research.
Collapse
|
6
|
Cooper DD, Frenguelli BG. The influence of sensory experience on the glutamatergic synapse. Neuropharmacology 2021; 193:108620. [PMID: 34048870 DOI: 10.1016/j.neuropharm.2021.108620] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 12/17/2022]
Abstract
The ability of glutamatergic synaptic strength to change in response to prevailing neuronal activity is believed to underlie the capacity of animals, including humans, to learn from experience. This learning better equips animals to safely navigate challenging and potentially harmful environments, while reinforcing behaviours that are conducive to survival. Early descriptions of the influence of experience on behaviour were provided by Donald Hebb who showed that an enriched environment improved performance of rats in a variety of behavioural tasks, challenging the widely-held view at the time that psychological development and intelligence were largely predetermined through genetic inheritance. Subsequent studies in a variety of species provided detailed cellular and molecular insights into the neurobiological adaptations associated with enrichment and its counterparts, isolation and deprivation. Here we review those experience-dependent changes that occur at the glutamatergic synapse, and which likely underlie the enhanced cognition associated with enrichment. We focus on the importance of signalling initiated by the release of BDNF and a prime downstream effector, MSK1, in orchestrating the many structural and functional neuronal adaptations associated with enrichment. In particular we discuss the MSK1-dependent expansion of the dynamic range of the glutamatergic synapse, which may allow enhanced information storage or processing, and the establishment of a genomic homeostasis that may both stabilise the enriched brain, and may make it better able to respond to novel experiences.
Collapse
Affiliation(s)
- Daniel D Cooper
- School of Life Sciences, University of Warwick, Coventry, UK
| | | |
Collapse
|
7
|
Morice E, Enderlin V, Gautron S, Laroche S. Contrasting Functions of Mitogen- and Stress-activated Protein Kinases 1 and 2 in Recognition Memory and In Vivo Hippocampal Synaptic Transmission. Neuroscience 2021; 463:70-85. [PMID: 33722673 DOI: 10.1016/j.neuroscience.2021.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 11/28/2022]
Abstract
The mitogen-activated protein kinases (MAPK) are major signaling components of intracellular pathways required for memory consolidation. Mitogen- and stress-activated protein kinases 1 and 2 (MSK1 and MSK2) mediate signal transduction downstream of MAPK. MSKs are activated by Extracellular-signal Regulated Kinase 1/2 (ERK1/2) and p38 MAPK. In turn, they can activate cyclic AMP-response-element-binding protein (CREB), thereby modulating the expression of immediate early genes crucial for the formation of long-term memories. While MSK1 has been previously implicated in certain forms of learning and memory, little is known concerning MSK2. Our goal was to explore the respective contribution of MSK1 and MSK2 in hippocampal synaptic transmission and plasticity and hippocampal-dependent recognition memory. In Msk1- and Msk2-knockout mice, we evaluated object and object-place recognition memory, basal synaptic transmission, paired-pulse facilitation (PPF) and inhibition (PPI), and the capacity to induce and sustain long-term potentiation (LTP) in vivo. We also assessed the level of two proteins downstream in the MAPK/ERK1/2 pathway crucial for long-term memory, CREB and the immediate early gene (IEG) Early growth response 1 (EGR1). Loss of Msk1, but not of Msk2, affected excitatory synaptic transmission at perforant path-to-dentate granule cell synapses, altered short-term presynaptic plasticity, impaired selectively long-term spatial recognition memory, and decreased basal levels of CREB and its activated form. LTP in vivo and LTP-induced CREB phosphorylation and EGR1 expression were unchanged after Msk1 or Msk2 deletion. Our findings demonstrate a dissimilar contribution of MSKs proteins in cognitive processes and suggest that Msk1 loss-of-function only has a deleterious impact on neuronal activity and hippocampal-dependent memory consolidation.
Collapse
Affiliation(s)
- Elise Morice
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Institut de Biologie Paris Seine, 75005 Paris, France; University Paris-Saclay, CNRS, Paris-Saclay Neuroscience Institute, 91405 Orsay, France.
| | - Valérie Enderlin
- University Paris-Saclay, CNRS, Paris-Saclay Neuroscience Institute, 91405 Orsay, France.
| | - Sophie Gautron
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Institut de Biologie Paris Seine, 75005 Paris, France.
| | - Serge Laroche
- University Paris-Saclay, CNRS, Paris-Saclay Neuroscience Institute, 91405 Orsay, France.
| |
Collapse
|
8
|
Olateju OI, Morè L, Arthur JSC, Frenguelli BG. Mitogen and Stress-activated Protein Kinase 1 Negatively Regulates Hippocampal Neurogenesis. Neuroscience 2020; 452:228-234. [PMID: 33246062 PMCID: PMC7810160 DOI: 10.1016/j.neuroscience.2020.11.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 10/20/2020] [Accepted: 11/10/2020] [Indexed: 12/18/2022]
Abstract
Neurogenesis in the subgranular zone (SGZ) of the adult hippocampus can be stimulated by a variety of means, including via exposure of experimental animals to an enriched environment that provides additional sensory, social, and motor stimulation. Tangible health and cognitive benefits accrue in enriched animals, including the amelioration of signs modelling psychiatric, neurological and neurodegenerative conditions that affect humans, which may in part be due to enhanced production of neurons. A key factor in the neuronal response to enrichment is the release of brain-derived neurotrophic factor (BDNF) and the activation of the Mitogen-Activated Protein Kinase (MAPK) cascade, which can lead to the stimulation of neurogenesis. Mitogen- and Stress-Activated protein Kinase 1 (MSK1) is a nuclear enzyme downstream of BDNF and MAPK that regulates transcription. MSK1 has previously been implicated in both basal and stimulated neurogenesis on the basis of studies with mice lacking MSK1 protein. In the present study, using mice in which only the kinase activity of MSK1 is lacking, we show that the rate of cellular proliferation in the SGZ (Ki-67 staining) is unaffected by the MSK1 kinase-dead (KD) mutation, and no different from controls levels after five weeks of enrichment. However, compared to wild-type mice, the number of doublecortin (DCX)-positive cells was greater in both standard-housed and enriched MSK1 KD mice. These observations suggest that, while MSK1 does not influence the basal rate of proliferation of neuronal precursors, MSK1 negatively regulates the number of cells destined to become neurons, potentially as a homeostatic control on the number of new neurons integrating into the dentate gyrus.
Collapse
Affiliation(s)
- Oladiran I Olateju
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK; School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, South Africa
| | - Lorenzo Morè
- School of Pharmacy and Biomedical Sciences, College of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK
| | - J Simon C Arthur
- School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | | |
Collapse
|
9
|
Pereira PDC, Henrique EP, Porfírio DM, Crispim CCDS, Campos MTB, de Oliveira RM, Silva IMS, Guerreiro LCF, da Silva TWP, da Silva ADJF, Rosa JBDS, de Azevedo DLF, Lima CGC, Castro de Abreu C, Filho CS, Diniz DLWP, Magalhães NGDM, Guerreiro-Diniz C, Diniz CWP, Diniz DG. Environmental Enrichment Improved Learning and Memory, Increased Telencephalic Cell Proliferation, and Induced Differential Gene Expression in Colossoma macropomum. Front Pharmacol 2020; 11:840. [PMID: 32595498 PMCID: PMC7303308 DOI: 10.3389/fphar.2020.00840] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 05/21/2020] [Indexed: 01/06/2023] Open
Abstract
Fish use spatial cognition based on allocentric cues to navigate, but little is known about how environmental enrichment (EE) affects learning and memory in correlation with hematological changes or gene expression in the fish brain. Here we investigated these questions in Colossoma macropomum (Teleostei). Fish were housed for 192 days in either EE or in an impoverished environment (IE) aquarium. EE contained toys, natural plants, and a 12-h/day water stream for voluntary exercise, whereas IE had no toys, plants, or water stream. A third plus maze aquarium was used for spatial and object recognition tests. Compared with IE, the EE fish showed greater learning rates, body length, and body weight. After behavioral tests, whole brain tissue was taken, stored in RNA-later, and then homogenized for DNA sequencing after conversion of isolated RNA. To compare read mapping and gene expression profiles across libraries for neurotranscriptome differential expression, we mapped back RNA-seq reads to the C. macropomum de novo assembled transcriptome. The results showed significant differential behavior, cell counts and gene expression in EE and IE individuals. As compared with IE, we found a greater number of cells in the telencephalon of individuals maintained in EE but no significant difference in the tectum opticum, suggesting differential plasticity in these areas. A total of 107,669 transcripts were found that ultimately yielded 64 differentially expressed transcripts between IE and EE brains. Another group of adult fish growing in aquaculture conditions were either subjected to exercise using running water flow or maintained sedentary. Flow cytometry analysis of peripheral blood showed a significantly higher density of lymphocytes, and platelets but no significant differences in erythrocytes and granulocytes. Thus, under the influence of contrasting environments, our findings showed differential changes at the behavioral, cellular, and molecular levels. We propose that the differential expression of selected transcripts, number of telencephalic cell counts, learning and memory performance, and selective hematological cell changes may be part of Teleostei adaptive physiological responses triggered by EE visuospatial and somatomotor stimulation. Our findings suggest abundant differential gene expression changes depending on environment and provide a basis for exploring gene regulation mechanisms under EE in C. macropomum.
Collapse
Affiliation(s)
- Patrick Douglas Corrêa Pereira
- Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Ciência e Tecnologia do Pará, Bragança, Brazil
| | - Ediely Pereira Henrique
- Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Ciência e Tecnologia do Pará, Bragança, Brazil
| | - Danillo Monteiro Porfírio
- Laboratório de Investigação em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | | | - Maitê Thaís Barros Campos
- Laboratório de Investigação em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | - Renata Melo de Oliveira
- Laboratório de Investigação em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | - Isabella Mesquita Sfair Silva
- Laboratório de Investigação em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | - Luma Cristina Ferreira Guerreiro
- Laboratório de Investigação em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | - Tiago Werley Pires da Silva
- Laboratório de Investigação em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | | | - João Batista da Silva Rosa
- Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Ciência e Tecnologia do Pará, Bragança, Brazil
| | | | - Cecília Gabriella Coutinho Lima
- Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Ciência e Tecnologia do Pará, Bragança, Brazil
| | - Cintya Castro de Abreu
- Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Ciência e Tecnologia do Pará, Bragança, Brazil
| | - Carlos Santos Filho
- Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Ciência e Tecnologia do Pará, Bragança, Brazil
| | | | - Nara Gyzely de Morais Magalhães
- Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Ciência e Tecnologia do Pará, Bragança, Brazil
| | - Cristovam Guerreiro-Diniz
- Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Ciência e Tecnologia do Pará, Bragança, Brazil
| | - Cristovam Wanderley Picanço Diniz
- Laboratório de Investigação em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | - Daniel Guerreiro Diniz
- Laboratório de Investigação em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém, Brazil
| |
Collapse
|
10
|
Experience Recruits MSK1 to Expand the Dynamic Range of Synapses and Enhance Cognition. J Neurosci 2020; 40:4644-4660. [PMID: 32376781 PMCID: PMC7294801 DOI: 10.1523/jneurosci.2765-19.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 04/17/2020] [Accepted: 04/20/2020] [Indexed: 12/29/2022] Open
Abstract
Experience powerfully influences neuronal function and cognitive performance, but the cellular and molecular events underlying the experience-dependent enhancement of mental ability have remained elusive. In particular, the mechanisms that couple the external environment to the genomic changes underpinning this improvement are unknown. To address this, we have used male mice harboring an inactivating mutation of mitogen- and stress-activated protein kinase 1 (MSK1), a brain-derived neurotrophic factor (BDNF)-activated enzyme downstream of the mitogen-activated protein kinase (MAPK) pathway. We show that MSK1 is required for the full extent of experience-induced improvement of spatial memory, for the expansion of the dynamic range of synapses, exemplified by the enhancement of hippocampal long-term potentiation (LTP) and long-term depression (LTD), and for the regulation of the majority of genes influenced by enrichment. In addition, and unexpectedly, we show that experience is associated with an MSK1-dependent downregulation of key MAPK and plasticity-related genes, notably of EGR1/Zif268 and Arc/Arg3.1, suggesting the establishment of a novel genomic landscape adapted to experience. By coupling experience to homeostatic changes in gene expression MSK1, represents a prime mechanism through which the external environment has an enduring influence on gene expression, synaptic function, and cognition. SIGNIFICANCE STATEMENT Our everyday experiences strongly influence the structure and function of the brain. Positive experiences encourage the growth and development of the brain and support enhanced learning and memory and resistance to mood disorders such as anxiety. While this has been known for many years, how this occurs is not clear. Here, we show that many of the positive aspects of experience depend on an enzyme called mitogen- and stress-activated protein kinase 1 (MSK1). Using male mice with a mutation in MSK1, we show that MSK1 is necessary for the majority of gene expression changes associated with experience, extending the range over which the communication between neurons occurs, and for both the persistence of memory and the ability to learn new task rules.
Collapse
|
11
|
Shaw A, Arnold LD, Privitera L, Whitfield PD, Doherty MK, Morè L. A proteomic signature for CNS adaptations to the valence of environmental stimulation. Behav Brain Res 2020; 383:112515. [PMID: 32006564 DOI: 10.1016/j.bbr.2020.112515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/11/2020] [Accepted: 01/27/2020] [Indexed: 10/25/2022]
Abstract
Environmental Enrichment leads to a significant improvement in long-term performance across a range of cognitive functions in mammals and it has been shown to produce an increased synaptic density and neurogenesis. Nevertheless it is still an open question as to whether some key aspects of spatial learning & memory and procedural learning might be embodied by different molecular pathways to those of social cognition. Associated with synaptic changes and potentially underlying conditions, the Ras-ERK pathway has been proposed to be the primary mediator of in vivo adaptations to environmental enrichment, acting via the downstream Ras-ERK signalling kinase MSK1 and the transcription factor CREB. Herein, we show that valence of environmental stimulation increased social competition and that this is associated with a specific proteomic signature in the frontal lobe but notably not in the hippocampus. Specifically, we show that altering the valence of environmental stimuli affected the level of social competition, with mice from negatively enriched environments winning significantly more encounters-even though mice from positive were bigger and should display dominance. This behavioural phenotype was accompanied by changes in the proteome of the fronto-ventral pole of the brain, with a differential increase in the relative abundance of proteins involved in the mitochondrial metabolic processes of the TCA cycle and respiratory processes. Investigation of this proteomic signature may pave the way for the elucidation of novel pathways underpinning the behavioural changes caused by negative enrichment and further out understanding of conditions whose core feature is increased social competition.
Collapse
Affiliation(s)
- Andrew Shaw
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, PR1 2HE, UK
| | - Luke D Arnold
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, PR1 2HE, UK
| | - Lucia Privitera
- Centre for Discovery Brain Sciences, Edinburgh, EH8 9JZ, UK & School of Medicine, University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK
| | - Phillip D Whitfield
- Division of Biomedical Science, University of the Highlands and Islands, Inverness, IV2 3JH, UK
| | - Mary K Doherty
- Division of Biomedical Science, University of the Highlands and Islands, Inverness, IV2 3JH, UK
| | - Lorenzo Morè
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, PR1 2HE, UK.
| |
Collapse
|
12
|
Morè L, Lauterborn JC, Papaleo F, Brambilla R. Enhancing cognition through pharmacological and environmental interventions: Examples from preclinical models of neurodevelopmental disorders. Neurosci Biobehav Rev 2020; 110:28-45. [PMID: 30981451 DOI: 10.1016/j.neubiorev.2019.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 12/29/2022]
Abstract
In this review we discuss the role of environmental and pharmacological treatments to enhance cognition with special regards to neurodevelopmental related disorders and aging. How the environment influences brain structure and function, and the interactions between rearing conditions and gene expression, are fundamental questions that are still poorly understood. We propose a model that can explain some of the discrepancies in findings for effects of environmental enrichment on outcome measures. Evidence of a direct causal correlation of nootropics and treatments that enhanced cognition also will be presented, and possible molecular mechanisms that include neurotrophin signaling and downstream pathways underlying these processes are discussed. Finally we review recent findings achieved with a wide set of behavioral and cognitive tasks that have translational validity to humans, and should be useful for future work on devising appropriate therapies. As will be discussed, the collective findings suggest that a combinational therapeutic approach of environmental enrichment and nootropics could be particularly successful for improving learning and memory in both developmental disorders and normal aging.
Collapse
Affiliation(s)
- Lorenzo Morè
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, PR1 2XT, Preston, UK.
| | - Julie C Lauterborn
- Department of Anatomy & Neurobiology, School of Medicine, University of California, Irvine, CA, 92617, USA.
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Istituto Italiano di Tecnologia, Via Morego, 30, 16163, Genova, Italy.
| | - Riccardo Brambilla
- Neuroscience and Mental Health Research Institute (NMHRI), Division of Neuroscience, School of Biosciences, Cardiff University, CF24 4HQ, Cardiff, UK.
| |
Collapse
|
13
|
McAllister BB, Thackray SE, de la Orta BKG, Gosse E, Tak P, Chipak C, Rehal S, Valverde Rascón A, Dyck RH. Effects of enriched housing on the neuronal morphology of mice that lack zinc transporter 3 (ZnT3) and vesicular zinc. Behav Brain Res 2019; 379:112336. [PMID: 31689442 DOI: 10.1016/j.bbr.2019.112336] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/10/2019] [Accepted: 10/28/2019] [Indexed: 12/19/2022]
Abstract
In the central nervous system, certain neurons store zinc within the synaptic vesicles of their axon terminals. This vesicular zinc can then be released in an activity-dependent fashion as an intercellular signal. The functions of vesicular zinc are not entirely understood, but evidence suggests that it is important for some forms of experience-dependent plasticity in the brain. The ability of neurons to store and release vesicular zinc is dependent on expression of the vesicular zinc transporter, ZnT3. Here, we examined the neuronal morphology of mice that lack ZnT3. Brains were collected from mice housed under standard laboratory conditions and from mice housed in enriched environments - large, multilevel enclosures with running wheels, numerous objects and tunnels, and a greater number of cage mates. Golgi-Cox staining was used to visualize neurons for analysis of dendritic length and dendritic spine density. Neurons were analyzed from the barrel cortex, striatum, basolateral amygdala, and hippocampus (CA1). ZnT3 knockout mice, relative to wild type mice, exhibited increased basal dendritic length in the layer 2/3 pyramidal neurons of barrel cortex, independently of housing condition. Environmental enrichment decreased apical dendritic length in these same neurons and increased dendritic spine density on striatal medium spiny neurons. Elimination of ZnT3 did not modulate any of the effects of enrichment. Our results provide no evidence that vesicular zinc is required for the experience-dependent changes that occur in response to environmental enrichment. They are consistent, however, with recent reports suggesting increased cortical volume in ZnT3 knockout mice.
Collapse
Affiliation(s)
- Brendan B McAllister
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Sarah E Thackray
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Brenda Karina Garciá de la Orta
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Elise Gosse
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Purnoor Tak
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Colten Chipak
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Sukhjinder Rehal
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Abril Valverde Rascón
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Richard H Dyck
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada.
| |
Collapse
|
14
|
Kheiri G, Dolatshahi M, Rahmani F, Rezaei N. Role of p38/MAPKs in Alzheimer's disease: implications for amyloid beta toxicity targeted therapy. Rev Neurosci 2019; 30:9-30. [PMID: 29804103 DOI: 10.1515/revneuro-2018-0008] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 03/22/2018] [Indexed: 01/06/2023]
Abstract
A myriad of environmental and genetic factors, as well as the physiologic process of aging, contribute to Alzheimer's disease (AD) pathology. Neuroinflammation is and has been a focus of interest, as a common gateway for initiation of many of the underlying pathologies of AD. Amyloid beta (Aβ) toxicity, increasing RAGE expression, tau hyperphosphorylation, induction of apoptosis, and deregulated autophagy are among other mechanisms, partly entangled and being explained by activation of mitogen-activated protein kinase (MAPK) and MAPK signaling. p38 MAPK is the most essential regulator of Aβ induced toxicity from this family. p38 induces NF-κB activation, glutamate excitotoxicity, and disruption of synaptic plasticity, which are other implications of all justifying the p38 MAPK as a potential target to break the vicious Aβ toxicity cycle. Until recently, many in vivo and in vitro studies have investigated the effects of p38 MAPK inhibitors in AD. The pyridinyl imidazole compounds SB202190 and SB203580 have shown promising anti-apoptotic results in vivo. MW108 inhibits activation of p38 and is able to postpone cognitive decline in animal models. The PD169316, with anti-inflammatory, anti-oxidative, and anti-apoptotic features, has improved spatial memory in vivo. Natural compounds from Camellia sinensis (green tea), polyphenols from olive oil, pinocembrin from propolis, and the puerarine extract isoflavones, have shown strong anti-apoptotic features, mediated by p38 MAPK inhibition. Use of these drug targets is limited due to central nervous system side effects or cross-reactivity with other kinases, predicting the low efficacy of these drugs in clinical trials.
Collapse
Affiliation(s)
- Ghazaleh Kheiri
- Student's Scientific Research Center (SSRC), Tehran University of Medical Sciences, 1416753955 Tehran, Iran.,NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), 19166 Tehran, Iran
| | - Mahsa Dolatshahi
- Student's Scientific Research Center (SSRC), Tehran University of Medical Sciences, 1416753955 Tehran, Iran.,NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), 19166 Tehran, Iran
| | - Farzaneh Rahmani
- Student's Scientific Research Center (SSRC), Tehran University of Medical Sciences, 1416753955 Tehran, Iran.,NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), 19166 Tehran, Iran
| | - Nima Rezaei
- NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), 19166 Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran 14194, Iran
| |
Collapse
|
15
|
Llorens-Martín M. Exercising New Neurons to Vanquish Alzheimer Disease. Brain Plast 2018; 4:111-126. [PMID: 30564550 PMCID: PMC6296267 DOI: 10.3233/bpl-180065] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2018] [Indexed: 02/07/2023] Open
Abstract
Alzheimer disease (AD) is the most common type of dementia in individuals over 65 years of age. The neuropathological hallmarks of the condition are Tau neurofibrillary tangles and Amyloid-β senile plaques. Moreover, certain susceptible regions of the brain experience a generalized lack of neural plasticity and marked synaptic alterations during the progression of this as yet incurable disease. One of these regions, the hippocampus, is characterized by the continuous addition of new neurons throughout life. This phenomenon, named adult hippocampal neurogenesis (AHN), provides a potentially endless source of new synaptic elements that increase the complexity and plasticity of the hippocampal circuitry. Numerous lines of evidence show that physical activity and environmental enrichment (EE) are among the most potent positive regulators of AHN. Given that neural plasticity is markedly decreased in many neurodegenerative diseases, the therapeutic potential of making certain lifestyle changes, such as increasing physical activity, is being recognised in several non-pharmacologic strategies seeking to slow down or prevent the progression of these diseases. This review article summarizes current evidence supporting the putative therapeutic potential of EE and physical exercise to increase AHN and hippocampal plasticity both under physiological and pathological circumstances, with a special emphasis on neurodegenerative diseases and AD.
Collapse
Affiliation(s)
- María Llorens-Martín
- Department of Molecular Neuropathology, Centro de Biología Molecular “Severo Ochoa”, CBMSO, CSIC-UAM, Madrid, Spain
- Center for Networked Biomedical Research on Neurodegenerative Diseases CIBERNED, Madrid, Spain
- Department of Molecular Biology, Faculty of Sciences, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
16
|
Wang H, Xu J, Lazarovici P, Quirion R, Zheng W. cAMP Response Element-Binding Protein (CREB): A Possible Signaling Molecule Link in the Pathophysiology of Schizophrenia. Front Mol Neurosci 2018; 11:255. [PMID: 30214393 PMCID: PMC6125665 DOI: 10.3389/fnmol.2018.00255] [Citation(s) in RCA: 285] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 07/06/2018] [Indexed: 12/17/2022] Open
Abstract
Dopamine is a brain neurotransmitter involved in the pathology of schizophrenia. The dopamine hypothesis states that, in schizophrenia, dopaminergic signal transduction is hyperactive. The cAMP-response element binding protein (CREB) is an intracellular protein that regulates the expression of genes that are important in dopaminergic neurons. Dopamine affects the phosphorylation of CREB via G protein-coupled receptors. Neurotrophins, such as brain derived growth factor (BDNF), are critical regulators during neurodevelopment and synaptic plasticity. The CREB is one of the major regulators of neurotrophin responses since phosphorylated CREB binds to a specific sequence in the promoter of BDNF and regulates its transcription. Moreover, susceptibility genes associated with schizophrenia also target and stimulate the activity of CREB. Abnormalities of CREB expression is observed in the brain of individuals suffering from schizophrenia, and two variants (-933T to C and -413G to A) were found only in schizophrenic patients. The CREB was also involved in the therapy of animal models of schizophrenia. Collectively, these findings suggest a link between CREB and the pathophysiology of schizophrenia. This review provides an overview of CREB structure, expression, and biological functions in the brain and its interaction with dopamine signaling, neurotrophins, and susceptibility genes for schizophrenia. Animal models in which CREB function is modulated, by either overexpression of the protein or knocked down through gene deletion/mutation, implicating CREB in schizophrenia and antipsychotic drugs efficacy are also discussed. Targeting research and drug development on CREB could potentially accelerate the development of novel medications against schizophrenia.
Collapse
Affiliation(s)
- Haitao Wang
- Department of Neuropharmacology and Drug Discovery, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jiangping Xu
- Department of Neuropharmacology and Drug Discovery, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Philip Lazarovici
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Remi Quirion
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Wenhua Zheng
- Faculty of Health Sciences, University of Macau, Taipa, China
| |
Collapse
|
17
|
Choi YS, Horning P, Aten S, Karelina K, Alzate-Correa D, Arthur JSC, Hoyt KR, Obrietan K. Mitogen- and Stress-Activated Protein Kinase 1 Regulates Status Epilepticus-Evoked Cell Death in the Hippocampus. ASN Neuro 2018; 9:1759091417726607. [PMID: 28870089 PMCID: PMC5588809 DOI: 10.1177/1759091417726607] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Mitogen-activated protein kinase (MAPK) signaling has been implicated in a wide range of neuronal processes, including development, plasticity, and viability. One of the principal downstream targets of both the extracellular signal-regulated kinase/MAPK pathway and the p38 MAPK pathway is Mitogen- and Stress-activated protein Kinase 1 (MSK1). Here, we sought to understand the role that MSK1 plays in neuroprotection against excitotoxic stimulation in the hippocampus. To this end, we utilized immunohistochemical labeling, a MSK1 null mouse line, cell viability assays, and array-based profiling approaches. Initially, we show that MSK1 is broadly expressed within the major neuronal cell layers of the hippocampus and that status epilepticus drives acute induction of MSK1 activation. In response to the status epilepticus paradigm, MSK1 KO mice exhibited a striking increase in vulnerability to pilocarpine-evoked cell death within the CA1 and CA3 cell layers. Further, cultured MSK1 null neurons exhibited a heighted level of N-methyl-D-aspartate-evoked excitotoxicity relative to wild-type neurons, as assessed using the lactate dehydrogenase assay. Given these findings, we examined the hippocampal transcriptional profile of MSK1 null mice. Affymetrix array profiling revealed that MSK1 deletion led to the significant (>1.25-fold) downregulation of 130 genes and an upregulation of 145 genes. Notably, functional analysis indicated that a subset of these genes contribute to neuroprotective signaling networks. Together, these data provide important new insights into the mechanism by which the MAPK/MSK1 signaling cassette confers neuroprotection against excitotoxic insults. Approaches designed to upregulate or mimic the functional effects of MSK1 may prove beneficial against an array of degenerative processes resulting from excitotoxic insults.
Collapse
Affiliation(s)
- Yun-Sik Choi
- 1 Department of Pharmaceutical Science and Technology, Catholic University of Daegu, Gyeongbuk, Republic of Korea
| | - Paul Horning
- 2 Department of Neuroscience, 2647 Ohio State University , Columbus, OH, USA
| | - Sydney Aten
- 2 Department of Neuroscience, 2647 Ohio State University , Columbus, OH, USA
| | - Kate Karelina
- 2 Department of Neuroscience, 2647 Ohio State University , Columbus, OH, USA
| | | | - J Simon C Arthur
- 4 College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, UK
| | - Kari R Hoyt
- 3 Division of Pharmacology, 2647 Ohio State University , Columbus, OH, USA
| | - Karl Obrietan
- 2 Department of Neuroscience, 2647 Ohio State University , Columbus, OH, USA
| |
Collapse
|
18
|
Hunter CJ, Remenyi J, Correa SA, Privitera L, Reyskens KMSE, Martin KJ, Toth R, Frenguelli BG, Arthur JSC. MSK1 regulates transcriptional induction of Arc/Arg3.1 in response to neurotrophins. FEBS Open Bio 2017; 7:821-834. [PMID: 28593137 PMCID: PMC5458472 DOI: 10.1002/2211-5463.12232] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/23/2017] [Accepted: 03/31/2017] [Indexed: 12/29/2022] Open
Abstract
The immediate early gene activity‐regulated cytoskeletal protein (Arc)/Arg3.1 and the neurotrophin brain‐derived neurotrophic factor (BDNF) play important roles in synaptic plasticity and learning and memory in the mammalian brain. However, the mechanisms by which BDNF regulates the expression of Arc/Arg3.1 are unclear. In this study, we show that BDNF acts via the ERK1/2 pathway to activate the nuclear kinase mitogen‐ and stress‐activated protein kinase 1 (MSK1). MSK1 then induces Arc/Arg3.1 expression via the phosphorylation of histone H3 at the Arc/Arg3.1 promoter. MSK1 can also phosphorylate the transcription factor cyclic‐AMP response element‐binding protein (CREB) on Ser133. However, this is not required for BDNF‐induced Arc.Arg3.1 transcription as a Ser133Ala knockin mutation had no effect on Arc/Arg3.1 induction. In parallel, ERK1/2 directly activates Arc/Arg3.1 mRNA transcription via at least one serum response element on the promoter, which bind a complex of the Serum Response Factor (SRF) and a Ternary Complex Factor (TCF).
Collapse
Affiliation(s)
- Chris J Hunter
- MRC Protein Phosphorylation Unit College of Life Sciences Sir James Black Centre University of Dundee UK
| | - Judit Remenyi
- Wellcome Trust Centre for Gene Regulation and Expression Wellcome Trust Building College of Life Sciences University of Dundee UK
| | - Sonia A Correa
- Bradford School of Pharmacy Faculty of Life Sciences University of Bradford UK
| | | | - Kathleen M S E Reyskens
- Division of Cell Signalling and Immunology Wellcome Trust Building College of Life Sciences University of Dundee UK
| | - Kirsty J Martin
- MRC Protein Phosphorylation Unit College of Life Sciences Sir James Black Centre University of Dundee UK
| | - Rachel Toth
- MRC Protein Phosphorylation Unit College of Life Sciences Sir James Black Centre University of Dundee UK
| | | | - J Simon C Arthur
- Division of Cell Signalling and Immunology Wellcome Trust Building College of Life Sciences University of Dundee UK
| |
Collapse
|
19
|
The Kinase Function of MSK1 Regulates BDNF Signaling to CREB and Basal Synaptic Transmission, But Is Not Required for Hippocampal Long-Term Potentiation or Spatial Memory. eNeuro 2017; 4:eN-NWR-0212-16. [PMID: 28275711 PMCID: PMC5318545 DOI: 10.1523/eneuro.0212-16.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 01/06/2017] [Accepted: 01/09/2017] [Indexed: 12/26/2022] Open
Abstract
The later stages of long-term potentiation (LTP) in vitro and spatial memory in vivo are believed to depend upon gene transcription. Accordingly, considerable attempts have been made to identify both the mechanisms by which transcription is regulated and indeed the gene products themselves. Previous studies have shown that deletion of one regulator of transcription, the mitogen- and stress-activated kinase 1 (MSK1), causes an impairment of spatial memory. Given the ability of MSK1 to regulate gene expression via the phosphorylation of cAMP response element binding protein (CREB) at serine 133 (S133), MSK1 is a plausible candidate as a prime regulator of transcription underpinning synaptic plasticity and learning and memory. Indeed, prior work has revealed the necessity for MSK1 in homeostatic and experience-dependent synaptic plasticity. However, using a knock-in kinase-dead mouse mutant of MSK1, the current study demonstrates that, while the kinase function of MSK1 is important in regulating the phosphorylation of CREB at S133 and basal synaptic transmission in hippocampal area CA1, it is not required for metabotropic glutamate receptor-dependent long-term depression (mGluR-LTD), two forms of LTP or several forms of spatial learning in the watermaze. These data indicate that other functions of MSK1, such as a structural role for the whole enzyme, may explain previous observations of a role for MSK1 in learning and memory.
Collapse
|
20
|
Tochiki KK, Maiarú M, Norris C, Hunt SP, Géranton SM. The mitogen and stress-activated protein kinase 1 regulates the rapid epigenetic tagging of dorsal horn neurons and nocifensive behaviour. Pain 2016; 157:2594-2604. [PMID: 27482631 PMCID: PMC5065054 DOI: 10.1097/j.pain.0000000000000679] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 07/15/2016] [Accepted: 07/20/2016] [Indexed: 12/15/2022]
Abstract
Phosphorylation of histone H3 at serine 10 (p-H3S10) is a marker of active gene transcription. Using cognitive models of neural plasticity, p-H3S10 was shown to be downstream of extracellular signal-regulated kinase (ERK) signalling in the hippocampus. In this study, we show that nociceptive signalling after peripheral formalin injection increased p-H3S10 expression in the ipsilateral dorsal horn. This increase was maximal 30 minutes after formalin injection and occurred mainly within p-ERK-positive neurons. Spinal p-H3S10-enhanced expression was also observed in neurokinin 1 receptor (NK1R), c-Fos, and Zif268 positive neurons and was inhibited by ablation of serotonergic descending controls. The mitogen and stress-activated protein kinase 1 (MSK1) is downstream of ERK and can induce p-H3S10. We found that, after formalin injection, most phospho-MSK1 (p-MSK1)-positive cells (87% ± 3%) expressed p-ERK and the majority of p-H3S10-positive cells (85% ± 5%) expressed p-MSK1. Inhibition of ERK activity with the MEK inhibitor SL327 reduced formalin-induced p-ERK, p-MSK1, and p-H3S10, demonstrating that spinal p-MSK1 and p-H3S10 were at least partly downstream of ERK signalling. Crucially, pharmacological blockade of spinal MSK1 activity with the novel MSK1 inhibitor SB727651A inhibited formalin-induced spinal p-H3S10 and nocifensive behaviour. These findings are the first to establish the involvement of p-H3S10 and its main kinase, MSK1, in ERK regulation of nociception. Given the general importance of ERK signalling in pain processing, our results suggest that p-H3S10 could play a role in the response to injury.
Collapse
Affiliation(s)
- Keri K. Tochiki
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Maria Maiarú
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Caspar Norris
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Stephen P. Hunt
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Sandrine M. Géranton
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| |
Collapse
|
21
|
Wu KL, Wu CW, Tain YL, Huang LT, Chao YM, Hung CY, Wu JC, Chen SR, Tsai PC, Chan JY. Environmental stimulation rescues maternal high fructose intake-impaired learning and memory in female offspring: Its correlation with redistribution of histone deacetylase 4. Neurobiol Learn Mem 2016; 130:105-17. [DOI: 10.1016/j.nlm.2016.02.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 01/26/2016] [Accepted: 02/05/2016] [Indexed: 12/12/2022]
|
22
|
Garthe A, Roeder I, Kempermann G. Mice in an enriched environment learn more flexibly because of adult hippocampal neurogenesis. Hippocampus 2015; 26:261-71. [PMID: 26311488 PMCID: PMC5049654 DOI: 10.1002/hipo.22520] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 08/25/2015] [Indexed: 12/21/2022]
Abstract
We here show that living in a stimulus‐rich environment (ENR) improves water maze learning with respect to specific key indicators that in previous loss‐of‐function experiments have been shown to rely on adult hippocampal neurogenesis. Analyzing the strategies employed by mice to locate the hidden platform in the water maze revealed that ENR facilitated task acquisition by increasing the probability to use effective search strategies. ENR also enhanced the animals’ behavioral flexibility, when the escape platform was moved to a new location. Treatment with temozolomide, which is known to reduce adult neurogenesis, abolished the effects of ENR on both acquisition and flexibility, while leaving other aspects of water maze learning untouched. These characteristic effects and interdependencies were not seen in parallel experiments with voluntary wheel running (RUN), a second pro‐neurogenic behavioral stimulus. Since the histological assessment of adult neurogenesis is by necessity an end‐point measure, the levels of neurogenesis over the course of the experiment can only be inferred and the present study focused on behavioral parameters as analytical endpoints. Although the correlation of physical activity with precursor cell proliferation and of learning and the survival of new neurons is well established, how the specific functional effects described here relate to dynamic changes in the stem cell niche remains to be addressed. Nevertheless, our findings support the hypothesis that adult neurogenesis is a critical mechanism underlying the beneficial effects of leading an active live, rich in experiences. © 2015 The Authors Hippocampus Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Alexander Garthe
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Adult Neurogenesis, Dresden, Germany.,CRTD-DFG Research Center for Regenerative Therapies Dresden, Genomics of Regeneration, Technische Universität Dresden, Dresden, Germany
| | - Ingo Roeder
- Institute for Medical Informatics and Biometry, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Gerd Kempermann
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Adult Neurogenesis, Dresden, Germany.,CRTD-DFG Research Center for Regenerative Therapies Dresden, Genomics of Regeneration, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
23
|
Loss CM, Binder LB, Muccini E, Martins WC, de Oliveira PA, Vandresen-Filho S, Prediger RD, Tasca CI, Zimmer ER, Costa-Schmidt LE, de Oliveira DL, Viola GG. Influence of environmental enrichment vs. time-of-day on behavioral repertoire of male albino Swiss mice. Neurobiol Learn Mem 2015; 125:63-72. [PMID: 26247375 DOI: 10.1016/j.nlm.2015.07.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 06/30/2015] [Accepted: 07/24/2015] [Indexed: 11/19/2022]
Abstract
Environmental enrichment (EE) is a non-pharmacological manipulation that promotes diverse forms of benefits in the central nervous system of captive animals. It is thought that EE influences animal behavior in a specie-(strain)-specific manner. Since rodents in general present different behaviors during distinct periods of the day, in this study we aimed to investigate the influence of time-of-day on behavioral repertoire of Swiss mice that reared in EE. Forty male Swiss mice (21days old) were housed in standard (SC) or enriched conditions (EC) for 60days. Behavioral assessments were conducted during the light phase (in presence of light) or dark phase (in absence of light) in the following tasks: open field, object recognition and elevated plus maze. First, we observed that the locomotor and exploratory activities are distinct between SC and EC groups only during the light phase. Second, we observed that "self-protective behaviors" were increased in EC group only when mice were tested during the light phase. However, "less defensive behaviors" were not affected by both housing conditions and time-of-day. Third, we showed that the performance of EE animals in object recognition task was improved in both light and dark conditions. Our findings highlight that EE-induced alterations in exploratory and emotional behaviors are just evident during light conditions. However, EE-induced cognitive benefits are remarkable even during dark conditions, when exploratory and emotional behaviors were similar between groups.
Collapse
Affiliation(s)
- Cássio Morais Loss
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Luisa Bandeira Binder
- Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
| | - Eduarda Muccini
- Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
| | - Wagner Carbolin Martins
- Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
| | | | - Samuel Vandresen-Filho
- Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Departamento de Ciências Básicas em Saúde, Faculdade de Medicina, Universidade Federal de Mato Grosso, Cuiabá, MT, Brazil.
| | - Rui Daniel Prediger
- Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
| | - Carla Inês Tasca
- Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
| | - Eduardo R Zimmer
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Luiz Ernesto Costa-Schmidt
- Laboratorio de Biología Reproductiva y Evolución, Instituto de Diversidad y Ecología Animal - IDEA/CONICET, Universidad Nacional de Córdoba - UNC, Vélez Sarsfield 299, 5000 Córdoba, Argentina; Programa de Pós-Graduação em Biologia, Universidade do Vale do Rio dos Sinos - UNISINOS, São Leopoldo, RS, Brazil.
| | - Diogo Losch de Oliveira
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Giordano Gubert Viola
- Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Universidade Federal de Santa Catarina, Campus Curitibanos, Curitibanos, SC, Brazil; Programa de Pós-Graduação em Ciências Fisiológicas, Centro de Ciências Biológica, Universidade Federal do Sergipe, São Cristóvão, SE, Brazil.
| |
Collapse
|
24
|
Aimone JB, Li Y, Lee SW, Clemenson GD, Deng W, Gage FH. Regulation and function of adult neurogenesis: from genes to cognition. Physiol Rev 2014; 94:991-1026. [PMID: 25287858 DOI: 10.1152/physrev.00004.2014] [Citation(s) in RCA: 441] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Adult neurogenesis in the hippocampus is a notable process due not only to its uniqueness and potential impact on cognition but also to its localized vertical integration of different scales of neuroscience, ranging from molecular and cellular biology to behavior. This review summarizes the recent research regarding the process of adult neurogenesis from these different perspectives, with particular emphasis on the differentiation and development of new neurons, the regulation of the process by extrinsic and intrinsic factors, and their ultimate function in the hippocampus circuit. Arising from a local neural stem cell population, new neurons progress through several stages of maturation, ultimately integrating into the adult dentate gyrus network. The increased appreciation of the full neurogenesis process, from genes and cells to behavior and cognition, makes neurogenesis both a unique case study for how scales in neuroscience can link together and suggests neurogenesis as a potential target for therapeutic intervention for a number of disorders.
Collapse
Affiliation(s)
- James B Aimone
- Cognitive Modeling Group, Sandia National Laboratories, Albuquerque, New Mexico; and Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, California
| | - Yan Li
- Cognitive Modeling Group, Sandia National Laboratories, Albuquerque, New Mexico; and Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, California
| | - Star W Lee
- Cognitive Modeling Group, Sandia National Laboratories, Albuquerque, New Mexico; and Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, California
| | - Gregory D Clemenson
- Cognitive Modeling Group, Sandia National Laboratories, Albuquerque, New Mexico; and Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, California
| | - Wei Deng
- Cognitive Modeling Group, Sandia National Laboratories, Albuquerque, New Mexico; and Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, California
| | - Fred H Gage
- Cognitive Modeling Group, Sandia National Laboratories, Albuquerque, New Mexico; and Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, California
| |
Collapse
|
25
|
Karelina K, Liu Y, Alzate-Correa D, Wheaton KL, Hoyt KR, Arthur JSC, Obrietan K. Mitogen and stress-activated kinases 1/2 regulate ischemia-induced hippocampal progenitor cell proliferation and neurogenesis. Neuroscience 2014; 285:292-302. [PMID: 25451279 DOI: 10.1016/j.neuroscience.2014.10.053] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 09/30/2014] [Accepted: 10/20/2014] [Indexed: 01/10/2023]
Abstract
Pathophysiological conditions such as cerebral ischemia trigger the production of new neurons from the neurogenic niche within the subgranular zone (SGZ) of the dentate gyrus. The functional significance of ischemia-induced neurogenesis is believed to be the regeneration of lost cells, thus contributing to post-ischemia recovery. However, the cell signaling mechanisms by which this process is regulated are still under investigation. Here, we investigated the role of mitogen and stress-activated protein kinases (MSK1/2) in the regulation of progenitor cell proliferation and neurogenesis after cerebral ischemia. Using the endothelin-1 model of ischemia, wild-type (WT) and MSK1(-/-)/MSK2(-/-) (MSK dKO) mice were injected with BrdU and sacrificed 2 days, 4 weeks, or 6 weeks later for the analysis of progenitor cell proliferation, neurogenesis, and neuronal morphology, respectively. We report a decrease in SGZ progenitor cell proliferation in MSK dKO mice compared to WT mice. Moreover, MSK dKO mice exhibited reduced neurogenesis and a delayed maturation of ischemia-induced newborn neurons. Further, structural analysis of neuronal arborization revealed reduced branching complexity in MSK dKO compared to WT mice. Taken together, this dataset suggests that MSK1/2 plays a significant role in the regulation of ischemia-induced progenitor cell proliferation and neurogenesis. Ultimately, revealing the cell signaling mechanisms that promote neuronal recovery will lead to novel pharmacological approaches for the treatment of neurodegenerative diseases such as cerebral ischemia.
Collapse
Affiliation(s)
- K Karelina
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Y Liu
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - D Alzate-Correa
- Division of Pharmacology, The Ohio State University, Columbus, OH 43210, USA
| | - K L Wheaton
- Division of Pharmacology, The Ohio State University, Columbus, OH 43210, USA
| | - K R Hoyt
- Division of Pharmacology, The Ohio State University, Columbus, OH 43210, USA
| | - J S C Arthur
- MRC Protein Phosphorylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | - K Obrietan
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
26
|
The roles of p38 MAPK/MSK1 signaling pathway in the neuroprotection of hypoxic postconditioning against transient global cerebral ischemia in adult rats. Mol Neurobiol 2013; 49:1338-49. [PMID: 24352802 DOI: 10.1007/s12035-013-8611-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 12/08/2013] [Indexed: 01/29/2023]
Abstract
Postconditioning has regenerated interest as a mechanical intervention against cerebral ischemia/reperfusion injury, but its molecular mechanisms remain unknown. We previously reported that hypoxic postconditioning (HPC) ameliorated neuronal death induced by transient global cerebral ischemia (tGCI) in hippocampal CA1 subregion of adult rats. This study tested the hypothesis that p38-mitogen-activated protein kinase (p38 MAPK)/mitogen- and stress-response kinase 1 (MSK1) signaling pathway plays a role in the HPC-induced neuroprotection. Male Wistar rats were subjected to 10 min ischemia induced by applying the four-vessel occlusion method. HPC with 120 min was applied at 24 h after reperfusion. Immunohistochemistry and Western blot were used to detect the expression of phosphorylation of p38 MAPK and MSK1, as well as cleaved caspase-3. We found that HPC induced a significant increase of phosphorylated p38 MAPK and MSK1 in neurons of hippocampal CA1 region and a significant decrease in glial cells after tGCI as well. Furthermore, HPC attenuated caspase-3 cleavation triggered by tGCI in CA1 region. Moreover, p38 MAPK inhibition by SB203580 significantly decreased the phosphorylation of MSK1, increased cleaved caspase-3 expression, and abolished the neuroprotection of HPC. These findings suggested that p38 MAPK/MSK1 signaling axis contributed to HPC-mediated neuroprotection against tGCI, at least in part, by regulating the activation of caspase-3.
Collapse
|
27
|
Ribosomal S6 kinase regulates ischemia-induced progenitor cell proliferation in the adult mouse hippocampus. Exp Neurol 2013; 253:72-81. [PMID: 24291236 DOI: 10.1016/j.expneurol.2013.11.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 10/14/2013] [Accepted: 11/22/2013] [Indexed: 12/14/2022]
Abstract
Ischemia-induced progenitor cell proliferation is a prominent example of the adult mammalian brain's ability to regenerate injured tissue resulting from pathophysiological processes. In order to better understand and exploit the cell signaling mechanisms that regulate ischemia-induced proliferation, we examined the role of the p42/44 mitogen-activated protein kinase (MAPK) cascade effector ribosomal S6 kinase (RSK) in this process. Here, using the endothelin-1 ischemia model in wild type mice, we show that the activated form of RSK is expressed in the progenitor cells of the subgranular zone (SGZ) after intrahippocampal cerebral ischemia. Further, RSK inhibition significantly reduces ischemia-induced SGZ progenitor cell proliferation. Using the neurosphere assay, we also show that both SGZ- and subventricular zone (SVZ)-derived adult neural stem cells (NSC) exhibit a significant reduction in proliferation in the presence of RSK and MAPK inhibitors. Taken together, these data reveal RSK as a regulator of ischemia-induced progenitor cell proliferation, and as such, suggest potential therapeutic value may be gained by specifically targeting the regulation of RSK in the progenitor cell population of the SGZ.
Collapse
|
28
|
Choi YS, Karelina K, Alzate-Correa D, Hoyt KR, Impey S, Arthur JS, Obrietan K. Mitogen- and stress-activated kinases regulate progenitor cell proliferation and neuron development in the adult dentate gyrus. J Neurochem 2012; 123:676-88. [PMID: 23020821 PMCID: PMC3575744 DOI: 10.1111/jnc.12035] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 08/21/2012] [Accepted: 09/25/2012] [Indexed: 01/19/2023]
Abstract
The neurogenic niche within the subgranular zone (SGZ) of the dentate gyrus is a source of new neurons throughout life. Interestingly, SGZ proliferative capacity is regulated by both physiological and pathophysiological conditions. One outstanding question involves the molecular mechanisms that regulate both basal and inducible adult neurogenesis. Here, we examined the role of the MAPK-regulated kinases, mitogen- and stress-activated kinase (MSK)1 and MSK2. as regulators of dentate gyrus SGZ progenitor cell proliferation and neurogenesis. Under basal conditions, MSK1/2 null mice exhibited significantly reduced progenitor cell proliferation capacity and a corollary reduction in the number of doublecortin (DCX)-positive immature neurons. Strikingly, seizure-induced progenitor proliferation was totally blocked in MSK1/2 null mice. This blunting of cell proliferation in MSK1/2 null mice was partially reversed by forskolin infusion, indicating that the inducible proliferative capacity of the progenitor cell population was intact. Furthermore, in MSK1/2 null mice, DCX-positive immature neurons exhibited reduced neurite arborization. Together, these data reveal a critical role for MSK1/2 as regulators of both basal and activity-dependent progenitor cell proliferation and morphological maturation in the SGZ.
Collapse
Affiliation(s)
- Yun-Sik Choi
- Department of Pharmaceutical Science & Technology, Catholic University of Daegu, Gyeongbuk, Rep. of Korea
| | - Kate Karelina
- Department of Neuroscience, Ohio State University, Columbus, OH 43210
| | | | - Kari R. Hoyt
- Division of Pharmacology, Ohio State University, Columbus, OH 43210
| | - Soren Impey
- Department of Cell and Developmental Biology, Oregon Health & Sciences University, Portland, OR 97239
| | - J. Simon Arthur
- MRC Protein Phosphorylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, U.K
| | - Karl Obrietan
- Department of Neuroscience, Ohio State University, Columbus, OH 43210
| |
Collapse
|