1
|
Devine EA, Imami AS, Eby H, Sahay S, Hamoud AR, Golchin H, Ryan W, Shedroff EA, Arvay T, Joyce AW, Asah SM, Walss-Bass C, O'Donovan S, McCullumsmith RE. Neuronal alterations in AKT isotype expression in schizophrenia. Mol Psychiatry 2024:10.1038/s41380-024-02770-8. [PMID: 39424930 DOI: 10.1038/s41380-024-02770-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 09/05/2024] [Accepted: 09/25/2024] [Indexed: 10/21/2024]
Abstract
Schizophrenia is characterized by substantial alterations in brain function, and previous studies suggest insulin signaling pathways, particularly involving AKT, are implicated in the pathophysiology of the disorder. This study demonstrates elevated mRNA expression of AKT1-3 in neurons from schizophrenia subjects, contrary to unchanged or diminished total AKT protein expression reported in previous postmortem studies, suggesting a potential decoupling of transcript and protein levels. Sex-specific differential AKT activity was observed, indicating divergent roles in males and females with schizophrenia. Alongside AKT, upregulation of PDPK1, a critical component of the insulin signaling pathway, and several protein phosphatases known to regulate AKT were detected. Moreover, enhanced expression of the transcription factor FOXO1, a regulator of glucose metabolism, hints at possible compensatory mechanisms related to insulin signaling dysregulation. Findings were largely independent of antipsychotic medication use, suggesting inherent alterations in schizophrenia. These results highlight the significance of AKT and related signaling pathways in schizophrenia, proposing that these changes might represent a compensatory response to a primary defect of canonical insulin signaling pathways. This research underscores the need for a detailed understanding of these signaling pathways for the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Emily A Devine
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA.
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Ali S Imami
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Hunter Eby
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Smita Sahay
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Abdul-Rizaq Hamoud
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Hasti Golchin
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - William Ryan
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Elizabeth A Shedroff
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Taylen Arvay
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Alex W Joyce
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Sophie M Asah
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Consuelo Walss-Bass
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Sinead O'Donovan
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Robert E McCullumsmith
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
- Department of Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
- Neurosciences Institute, ProMedica, Toledo, OH, USA
| |
Collapse
|
2
|
Devine EA, Imami AS, Eby H, Hamoud AR, Golchin H, Ryan W, Sahay S, Shedroff EA, Arvay T, Joyce AW, Asah SM, Walss-Bass C, O'Donovan S, McCullumsmith RE. Neuronal alterations in AKT isotype expression in schizophrenia. RESEARCH SQUARE 2024:rs.3.rs-3940448. [PMID: 38559131 PMCID: PMC10980160 DOI: 10.21203/rs.3.rs-3940448/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Schizophrenia is characterized by substantial alterations in brain function, and previous studies suggest insulin signaling pathways, particularly involving AKT, are implicated in the pathophysiology of the disorder. This study demonstrates elevated mRNA expression of AKT1-3 in neurons from schizophrenia subjects, contrary to unchanged or diminished total AKT protein expression reported in previous postmortem studies, suggesting a potential decoupling of transcript and protein levels. Sex-specific differential AKT activity was observed, indicating divergent roles in males and females with schizophrenia. Alongside AKT, upregulation of PDPK1, a critical component of the insulin signaling pathway, and several protein phosphatases known to regulate AKT were detected. Moreover, enhanced expression of the transcription factor FOXO1, a regulator of glucose metabolism, hints at possible compensatory mechanisms related to insulin signaling dysregulation. Findings were largely independent of antipsychotic medication use, suggesting inherent alterations in schizophrenia. These results highlight the significance of AKT and related signaling pathways in schizophrenia, proposing that these changes might represent a compensatory response to a primary defect of conical insulin signaling pathways. This research underscores the need for a detailed understanding of these signaling pathways for the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Emily A Devine
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ali S Imami
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Hunter Eby
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Abdul-Rizaq Hamoud
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Hasti Golchin
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - William Ryan
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Smita Sahay
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Elizabeth A Shedroff
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Taylen Arvay
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Alex W Joyce
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Sophie M Asah
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Consuelo Walss-Bass
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Sinead O'Donovan
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Robert E McCullumsmith
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
- Neurosciences Institute, ProMedica, Toledo, OH, USA
- Department of Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| |
Collapse
|
3
|
Sowndharya S, Rajan KE. Environmental enrichment improves social isolation-induced memory impairment: The possible role of ITSN1-Reelin-AMPA receptor signaling pathway. PLoS One 2024; 19:e0294354. [PMID: 38241230 PMCID: PMC10798460 DOI: 10.1371/journal.pone.0294354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 10/30/2023] [Indexed: 01/21/2024] Open
Abstract
Environmental enrichment (EE) through combination of social and non-biological stimuli enhances activity-dependent synaptic plasticity and improves behavioural performance. Our earlier studies have suggested that EE resilience the stress induced depression/ anxiety-like behaviour in Indian field mice Mus booduga. This study was designed to test whether EE reverses the social isolation (SI) induced effect and improve memory. Field-caught mice M. booduga were subjected to behaviour test (Direct wild, DW), remaining animals were housed under SI for ten days and then housed for short-term at standard condition (STSC)/ long-term at standard condition (LTSC) or as group in EE cage. Subsequently, we have examined reference, working memory and expression of genes associated with synaptic plasticity. Our analysis have shown that EE reversed SI induced impairment in reference, working memory and other accompanied changes i.e. increased level of Intersectin 1 (ITSN1), Huntingtin (Htt), Synaptotagmin -IV (SYT4), variants of brain-derived neurotrophic factor (Bdnf - III), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor (GluR1) expression, and decreased variants of Bdnf (IV), BDNF, Reelin, Apolipoprotein E receptor 2 (ApoER2), very low-density lipoprotein receptor (VLDLR), Src family tyrosine kinase (SFKs), Disabled protein (Dab)-1, Protein kinase B (PKB/Akt), GluR2, Mitogen-activated protein kinase (MAPK) and Extracellular signal-regulated kinase (ERK1/2) expression. In addition, SI induced reduction in BDNF expressing neurons in dentate gyrus of hippocampus reversed by EE. Further, we found that SI decreases small neuro-active molecules such as Benzenedicarboxylic acid, and increases 2-Pregnene in the hippocampus and feces reversed by EE. Overall, this study demonstrated that EE is effectively reversed the SI induced memory impairment by potentially regulating the molecules associated with the ITSN1-Reelin-AMPA receptor pathway to increase synaptic plasticity.
Collapse
Affiliation(s)
- Swamynathan Sowndharya
- Behavioural Neuroscience Laboratory, Department of Animal Science, Bharathidasan University, Tiruchirappalli, India
| | - Koilmani Emmanuvel Rajan
- Behavioural Neuroscience Laboratory, Department of Animal Science, Bharathidasan University, Tiruchirappalli, India
| |
Collapse
|
4
|
Identification of Peripheral Blood miRNA Biomarkers in First-Episode Drug-Free Schizophrenia Patients Using Bioinformatics Strategy. Mol Neurobiol 2022; 59:4730-4746. [DOI: 10.1007/s12035-022-02878-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 05/12/2022] [Indexed: 11/26/2022]
|
5
|
de Oliveira Figueiredo EC, Bondiolotti BM, Laugeray A, Bezzi P. Synaptic Plasticity Dysfunctions in the Pathophysiology of 22q11 Deletion Syndrome: Is There a Role for Astrocytes? Int J Mol Sci 2022; 23:ijms23084412. [PMID: 35457231 PMCID: PMC9028090 DOI: 10.3390/ijms23084412] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 01/01/2023] Open
Abstract
The 22q11 deletion syndrome (DS) is the most common microdeletion syndrome in humans and gives a high probability of developing psychiatric disorders. Synaptic and neuronal malfunctions appear to be at the core of the symptoms presented by patients. In fact, it has long been suggested that the behavioural and cognitive impairments observed in 22q11DS are probably due to alterations in the mechanisms regulating synaptic function and plasticity. Often, synaptic changes are related to structural and functional changes observed in patients with cognitive dysfunctions, therefore suggesting that synaptic plasticity has a crucial role in the pathophysiology of the syndrome. Most interestingly, among the genes deleted in 22q11DS, six encode for mitochondrial proteins that, in mouse models, are highly expressed just after birth, when active synaptogenesis occurs, therefore indicating that mitochondrial processes are strictly related to synapse formation and maintenance of a correct synaptic signalling. Because correct synaptic functioning, not only requires correct neuronal function and metabolism, but also needs the active contribution of astrocytes, we summarize in this review recent studies showing the involvement of synaptic plasticity in the pathophysiology of 22q11DS and we discuss the relevance of mitochondria in these processes and the possible involvement of astrocytes.
Collapse
Affiliation(s)
| | - Bianca Maria Bondiolotti
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland; (E.C.d.O.F.); (B.M.B.); (A.L.)
| | - Anthony Laugeray
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland; (E.C.d.O.F.); (B.M.B.); (A.L.)
| | - Paola Bezzi
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland; (E.C.d.O.F.); (B.M.B.); (A.L.)
- Department of Pharmacology and Physiology, University of Rome Sapienza, 00185 Rome, Italy
- Correspondence: or
| |
Collapse
|
6
|
Fei E, Chen P, Zhang Q, Zhong Y, Zhou T. Protein kinase B/Akt1 phosphorylates dysbindin-1A at serine 10 to regulate neuronal development. Neuroscience 2022; 490:66-78. [DOI: 10.1016/j.neuroscience.2022.01.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 01/24/2022] [Accepted: 01/28/2022] [Indexed: 01/05/2023]
|
7
|
Querfurth H, Marshall J, Parang K, Rioult-Pedotti MS, Tiwari R, Kwon B, Reisinger S, Lee HK. A PDK-1 allosteric agonist neutralizes insulin signaling derangements and beta-amyloid toxicity in neuronal cells and in vitro. PLoS One 2022; 17:e0261696. [PMID: 35061720 PMCID: PMC8782417 DOI: 10.1371/journal.pone.0261696] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 12/08/2021] [Indexed: 01/09/2023] Open
Abstract
The Alzheimer's brain is affected by multiple pathophysiological processes, which include a unique, organ-specific form of insulin resistance that begins early in its course. An additional complexity arises from the four-fold risk of Alzheimer's Disease (AD) in type 2 diabetics, however there is no definitive proof of causation. Several strategies to improve brain insulin signaling have been proposed and some have been clinically tested. We report findings on a small allosteric molecule that reverses several indices of insulin insensitivity in both cell culture and in vitro models of AD that emphasize the intracellular accumulation of β-amyloid (Aβi). PS48, a chlorophenyl pentenoic acid, is an allosteric activator of PDK-1, which is an Akt-kinase in the insulin/PI3K pathway. PS48 was active at 10 nM to 1 μM in restoring normal insulin-dependent Akt activation and in mitigating Aβi peptide toxicity. Synaptic plasticity (LTP) in prefrontal cortical slices from normal rat exposed to Aβ oligomers also benefited from PS48. During these experiments, neither overstimulation of PI3K/Akt signaling nor toxic effects on cells was observed. Another neurotoxicity model producing insulin insensitivity, utilizing palmitic acid, also responded to PS48 treatment, thus validating the target and indicating that its therapeutic potential may extend outside of β-amyloid reliance. The described in vitro and cell based-in vitro coupled enzymatic assay systems proved suitable platforms to screen a preliminary library of new analogs.
Collapse
Affiliation(s)
- Henry Querfurth
- Department of Neurology, Tufts Medical Center, Boston, MA, United States of America
| | - John Marshall
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI, United States of America
| | - Keykavous Parang
- Center for Targeted Drug Delivery, Chapman University, School of Pharmacology, Irvine, CA United States of America
| | - Mengia S. Rioult-Pedotti
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI, United States of America
- Department of Neurology, Clinical Neurorehabilitation, University of Zurich, Zurich, Switzerland
| | - Rakesh Tiwari
- Center for Targeted Drug Delivery, Chapman University, School of Pharmacology, Irvine, CA United States of America
| | - Bumsup Kwon
- Department of Neurology, Rhode Island Hospital, Providence, RI, United States of America
| | | | - Han-Kyu Lee
- Department of Neurology, Tufts Medical Center, Boston, MA, United States of America
| |
Collapse
|
8
|
Epigenetic Mediation of AKT1 rs1130233's Effect on Delta-9-Tetrahydrocannabinol-Induced Medial Temporal Function during Fear Processing. Brain Sci 2021; 11:brainsci11091240. [PMID: 34573260 PMCID: PMC8471665 DOI: 10.3390/brainsci11091240] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/29/2021] [Accepted: 07/29/2021] [Indexed: 12/28/2022] Open
Abstract
High doses of delta-9-tetrahydrocannabinol (THC), the main psychoactive component of cannabis, have been shown to have anxiogenic effects. Additionally, THC effects have been shown to be modulated by genotype, including the single nucleotide polymorphism (SNP) rs1130233 at the protein kinase AKT1 gene, a key component of the dopamine signalling cascade. As such, it is likely that epigenetic methylation around this SNP may affect AKT gene expression, which may in turn impact on the acute effects of THC on brain function. We investigated the genetic (AKT1 rs1130233) and epigenetic modulation of brain function during fear processing in a 2-session, double-blind, cross-over, randomized placebo-controlled THC administration, in 36 healthy males. Fear processing was assessed using an emotion (fear processing) paradigm, under functional magnetic resonance imaging (fMRI). Complete genetic and fMRI data were available for 34 participants. THC caused an increase in anxiety and transient psychotomimetic symptoms and para-hippocampal gyrus/amygdala activation. Number of A alleles at the AKT1 rs1130233 SNP, and percentage methylation at the CpG11-12 site, were independently associated with a greater effect of THC on activation in a network of brain regions including left and right parahippocampal gyri, respectively. AKT1 rs1130233 moderation of the THC effect on left parahippocampal activation persisted after covarying for methylation percentage, and was partially mediated in sections of the left parahippocampal gyrus/hippocampus by methylation percentage. These results may offer an example of how genetic and epigenetic variations influence the psychotomimetic and neurofunctional effects of THC.
Collapse
|
9
|
Mazuir E, Richevaux L, Nassar M, Robil N, de la Grange P, Lubetzki C, Fricker D, Sol-Foulon N. Oligodendrocyte Secreted Factors Shape Hippocampal GABAergic Neuron Transcriptome and Physiology. Cereb Cortex 2021; 31:5024-5041. [PMID: 34023893 DOI: 10.1093/cercor/bhab139] [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: 11/10/2020] [Revised: 03/26/2021] [Accepted: 04/17/2021] [Indexed: 11/14/2022] Open
Abstract
Oligodendrocytes form myelin for central nervous system axons and release factors which signal to neurons during myelination. Here, we ask how oligodendroglial factors influence hippocampal GABAergic neuron physiology. In mixed hippocampal cultures, GABAergic neurons fired action potentials (APs) of short duration and received high frequencies of excitatory synaptic events. In purified neuronal cultures without glial cells, GABAergic neuron excitability increased and the frequency of synaptic events decreased. These effects were largely reversed by adding oligodendrocyte conditioned medium (OCM). We compared the transcriptomic signature with the electrophysiological phenotype of single neurons in these three culture conditions. Genes expressed by single pyramidal or GABAergic neurons largely conformed to expected cell-type specific patterns. Multiple genes of GABAergic neurons were significantly downregulated by the transition from mixed cultures containing glial cells to purified neuronal cultures. Levels of these genes were restored by the addition of OCM to purified cultures. Clustering genes with similar changes in expression between different culture conditions revealed processes affected by oligodendroglial factors. Enriched genes are linked to roles in synapse assembly, AP generation, and transmembrane ion transport, including of zinc. These results provide new insight into the molecular targets by which oligodendrocytes influence neuron excitability and synaptic function.
Collapse
Affiliation(s)
- Elisa Mazuir
- Sorbonne University, Inserm, CNRS, Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, F-75013 Paris, France
| | | | - Merie Nassar
- Université de Paris, INCC UMR 8002, CNRS, F-75006 Paris
| | | | | | - Catherine Lubetzki
- Sorbonne University, Inserm, CNRS, Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, F-75013 Paris, France.,Assistance Publique des Hôpitaux de Paris (APHP), Neurology Department, Pitié-Salpêtrière hospital, Paris 75013, France
| | | | - Nathalie Sol-Foulon
- Sorbonne University, Inserm, CNRS, Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, F-75013 Paris, France
| |
Collapse
|
10
|
Palumbo S, Paterson C, Yang F, Hood VL, Law AJ. PKBβ/AKT2 deficiency impacts brain mTOR signaling, prefrontal cortical physiology, hippocampal plasticity and select murine behaviors. Mol Psychiatry 2021; 26:411-428. [PMID: 33328589 PMCID: PMC7854513 DOI: 10.1038/s41380-020-00964-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/31/2020] [Accepted: 11/16/2020] [Indexed: 12/11/2022]
Abstract
The serine/threonine protein kinase v-AKT homologs (AKTs), are implicated in typical and atypical neurodevelopment. Akt isoforms Akt1, Akt2, and Akt3 have been extensively studied outside the brain where their actions have been found to be complementary, non-overlapping and often divergent. While the neurological functions of Akt1 and Akt3 isoforms have been investigated, the role for Akt2 remains underinvestigated. Neurobehavioral, electrophysiological, morphological and biochemical assessment of Akt2 heterozygous and knockout genetic deletion in mouse, reveals a novel role for Akt2 in axonal development, dendritic patterning and cell-intrinsic and neural circuit physiology of the hippocampus and prefrontal cortex. Akt2 loss-of-function increased anxiety-like phenotypes, impaired fear conditioned learning, social behaviors and discrimination memory. Reduced sensitivity to amphetamine was observed, supporting a role for Akt2 in regulating dopaminergic tone. Biochemical analyses revealed dysregulated brain mTOR and GSK3β signaling, consistent with observed learning and memory impairments. Rescue of cognitive impairments was achieved through pharmacological enhancement of PI3K/AKT signaling and PIK3CD inhibition. Together these data highlight a novel role for Akt2 in neurodevelopment, learning and memory and show that Akt2 is a critical and non-redundant regulator of mTOR activity in brain.
Collapse
Affiliation(s)
- Sara Palumbo
- Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health Intramural Program, Bethesda MD 20892.,Department of Surgical, Medical and Molecular Pathology and Critical Care, University of Pisa, Pisa, Italy (current)
| | - Clare Paterson
- Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health Intramural Program, Bethesda MD 20892.,Department of Psychiatry, University of Colorado, School of Medicine. Aurora, CO 80045
| | - Feng Yang
- Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health Intramural Program, Bethesda MD 20892.,Division of Neurodegenerative Diseases and Translational Sciences Tiantan Hospital & Advanced Innovation Center for Human Brain Protection. Capital Medical University, Beijing, China (current)
| | - Veronica L. Hood
- Department of Psychiatry, University of Colorado, School of Medicine. Aurora, CO 80045
| | - Amanda J. Law
- Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health Intramural Program, Bethesda MD 20892.,Department of Psychiatry, University of Colorado, School of Medicine. Aurora, CO 80045.,To whom correspondence should be addressed:
| |
Collapse
|
11
|
The KBTBD6/7-DRD2 axis regulates pituitary adenoma sensitivity to dopamine agonist treatment. Acta Neuropathol 2020; 140:377-396. [PMID: 32572597 DOI: 10.1007/s00401-020-02180-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 06/12/2020] [Accepted: 06/12/2020] [Indexed: 12/19/2022]
Abstract
Pituitary adenoma (PA) is one of the most common intracranial tumors, and approximately 40% of all PAs are prolactinomas. Dopamine agonists (DAs), such as cabergoline (CAB), have been successfully used in the treatment of prolactinomas. The expression of dopamine type 2 receptor (DRD2) determines the therapeutic effect of DAs, but the molecular mechanisms of DRD2 regulation are not fully understood. In this study, we first demonstrated that DRD2 underwent proteasome-mediated degradation. We further employed the yeast two-hybrid system and identified kelch repeat and BTB (POZ) domain containing 7 (KBTBD7), a substrate adaptor for the CUL3-RING ubiquitin (Ub) ligase complex, as a DRD2-interacting protein. KBTBD6/7 directly interacted with, and ubiquitinated DRD2 at five ubiquitination sites (K221, K226, K241, K251, and K258). CAB, a high-affinity DRD2 agonist, induced DRD2 internalization, and cytoplasmic DRD2 was degraded via ubiquitination under the control of KBTBD6/7, the activity of which attenuated CAB-mediated inhibition of the AKT/mTOR pathway. KBTBD7 knockout (KO) mice were generated using the CRISPR-Cas9 technique, in which the static level of DRD2 protein was elevated in the pituitary gland, thalamus, and heart, compared to that of WT mice. Consistently, the expression of KBTBD6/7 was negatively correlated with that of DRD2 in human pituitary tumors. Moreover, KBTBD7 was highly expressed in dopamine-resistant prolactinomas, but at low levels in dopamine-sensitive prolactinomas. Knockdown of KBTBD6/7 sensitized MMQ cells and primary pituitary tumor cells to CAB treatment. Conversely, KBTBD7 overexpression increased CAB resistance of estrogen-induced in situ rat prolactinoma model. Together, our findings have uncovered the novel mechanism of DRD2 protein degradation and shown that the KBTBD6/7-DRD2 axis regulates PA sensitivity to DA treatment. KBTBD6/7 may thus become a promising therapeutic target for pituitary tumors.
Collapse
|
12
|
Ibarra-Lecue I, Diez-Alarcia R, Morentin B, Meana JJ, Callado LF, Urigüen L. Ribosomal Protein S6 Hypofunction in Postmortem Human Brain Links mTORC1-Dependent Signaling and Schizophrenia. Front Pharmacol 2020; 11:344. [PMID: 32265715 PMCID: PMC7105616 DOI: 10.3389/fphar.2020.00344] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/09/2020] [Indexed: 12/26/2022] Open
Abstract
The mechanistic target of rapamycin (also known as mammalian target of rapamycin) (mTOR)-dependent signaling pathway plays an important role in protein synthesis, cell growth, and proliferation, and has been linked to the development of the central nervous system. Recent studies suggest that mTOR signaling pathway dysfunction could be involved in the etiopathogenesis of schizophrenia. The main goal of this study was to evaluate the status of mTOR signaling pathway in postmortem prefrontal cortex (PFC) samples of subjects with schizophrenia. For this purpose, we quantified the protein expression and phosphorylation status of the mTOR downstream effector ribosomal protein S6 as well as other pathway interactors such as Akt and GSK3β. Furthermore, we quantified the status of these proteins in the brain cortex of rats chronically treated with the antipsychotics haloperidol, clozapine, or risperidone. We found a striking decrease in the expression of total S6 and in its active phosphorylated form phospho-S6 (Ser235/236) in the brain of subjects with schizophrenia compared to matched controls. The chronic treatment with the antipsychotics haloperidol and clozapine affected both the expression of GSK3β and the activation of Akt [phospho-Akt (Ser473)] in rat brain cortex, while no changes were observed in S6 and phospho-S6 (Ser235/236) protein expression with any antipsychotic treatment. These findings provide further evidence for the involvement of the mTOR-dependent signaling pathway in schizophrenia and suggest that a hypofunctional S6 may have a role in the etiopathogenesis of this disorder.
Collapse
Affiliation(s)
- Inés Ibarra-Lecue
- Department of Pharmacology, University of the Basque Country UPV/EHU and Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Leioa, Spain
| | - Rebeca Diez-Alarcia
- Department of Pharmacology, University of the Basque Country UPV/EHU and Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Leioa, Spain.,Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Benito Morentin
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.,Section of Forensic Pathology, Basque Institute of Legal Medicine, Bilbao, Spain
| | - J Javier Meana
- Department of Pharmacology, University of the Basque Country UPV/EHU and Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Leioa, Spain.,Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Luis F Callado
- Department of Pharmacology, University of the Basque Country UPV/EHU and Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Leioa, Spain.,Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Leyre Urigüen
- Department of Pharmacology, University of the Basque Country UPV/EHU and Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Leioa, Spain.,Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| |
Collapse
|
13
|
Howell KR, Law AJ. Neurodevelopmental concepts of schizophrenia in the genome-wide association era: AKT/mTOR signaling as a pathological mediator of genetic and environmental programming during development. Schizophr Res 2020; 217:95-104. [PMID: 31522868 PMCID: PMC7065975 DOI: 10.1016/j.schres.2019.08.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 08/28/2019] [Accepted: 08/31/2019] [Indexed: 12/14/2022]
Abstract
Normative brain development is contingent on the complex interplay between genes and environment. Schizophrenia (SCZ) is considered a highly polygenic, neurodevelopmental disorder associated with impaired neural circuit development, neurocognitive function and variations in neurotransmitter signaling systems, including dopamine. Significant evidence, accumulated over the last 30 years indicates a role for the in utero environment in SCZ pathophysiology. Emerging data suggests that changes in placental programming and function may mediate the link between genetic risk, early life complications (ELC) and adverse neurodevelopmental outcomes, with risk highlighted in key developmental drivers that converge on AKT/mTOR signaling. In this article we overview select risk genes identified through recent genome-wide association studies of SCZ including AKT3, miR-137, DRD2, and AKT1 itself. We propose that through convergence on AKT/mTOR signaling, these genes are critical factors directing both placentation and neurodevelopment, influencing risk for SCZ through dysregulation of placental function, metabolism and early brain development. We discuss association of risk genes in the context of their known roles in neurodevelopment, placental expression and their possible mechanistic links to SCZ in the broad context of the 'developmental origins of adult disease' construct. Understanding how common genetic variation impacts early fetal programming may advance our knowledge of disease etiology and identify early critical developmental windows for prevention and intervention.
Collapse
Affiliation(s)
| | - Amanda J. Law
- Corresponding Author: Amanda J. Law, PhD, Professor of Psychiatry, Medicine and Cell and Developmental Biology, Nancy L. Gary Endowed Chair in Children’s Mental Disorders Research, University of Colorado, School of Medicine, , Phone: 303-724-4418, Fax: 303-724-4425, 12700 E. 19th Ave., MS 8619, Aurora, CO 80045
| |
Collapse
|
14
|
Lithium for schizophrenia: supporting evidence from a 12-year, nationwide health insurance database and from Akt1-deficient mouse and cellular models. Sci Rep 2020; 10:647. [PMID: 31959776 PMCID: PMC6971245 DOI: 10.1038/s41598-019-57340-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022] Open
Abstract
Accumulating evidence suggests AKT1 and DRD2-AKT-GSK3 signaling involvement in schizophrenia. AKT1 activity is also required for lithium, a GSK3 inhibitor, to modulate mood-related behaviors. Notably, GSK3 inhibitor significantly alleviates behavioral deficits in Akt1−/− female mice, whereas typical/atypical antipsychotics have no effect. In agreement with adjunctive therapy with lithium in treating schizophrenia, our data mining indicated that the average utilization rates of lithium in the Taiwan National Health Insurance Research Database from 2002 to 2013 are 10.9% and 6.63% in inpatients and outpatients with schizophrenia, respectively. Given that lithium is commonly used in clinical practice, it is of great interest to evaluate the effect of lithium on alleviating Akt1-related deficits. Taking advantage of Akt1+/− mice to mimic genetic deficiency in patients, behavioral impairments were replicated in female Akt1+/− mice but were alleviated by subchronic lithium treatment for 13 days. Lithium also effectively alleviated the observed reduction in phosphorylated GSK3α/β expression in the brains of Akt1+/− mice. Furthermore, inhibition of Akt expression using an Akt1/2 inhibitor significantly reduced neurite length in P19 cells and primary hippocampal cell cultures, which was also ameliorated by lithium. Collectively, our findings implied the therapeutic potential of lithium and the importance of the AKT1-GSK3 signaling pathway.
Collapse
|
15
|
Ibarra-Lecue I, Diez-Alarcia R, Morentin B, Meana JJ, Callado LF, Urigüen L. Ribosomal Protein S6 Hypofunction in Postmortem Human Brain Links mTORC1-Dependent Signaling and Schizophrenia. Front Pharmacol 2020; 11:344. [PMID: 32265715 DOI: 10.3389/fphar.2020.00344/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/09/2020] [Indexed: 05/20/2023] Open
Abstract
The mechanistic target of rapamycin (also known as mammalian target of rapamycin) (mTOR)-dependent signaling pathway plays an important role in protein synthesis, cell growth, and proliferation, and has been linked to the development of the central nervous system. Recent studies suggest that mTOR signaling pathway dysfunction could be involved in the etiopathogenesis of schizophrenia. The main goal of this study was to evaluate the status of mTOR signaling pathway in postmortem prefrontal cortex (PFC) samples of subjects with schizophrenia. For this purpose, we quantified the protein expression and phosphorylation status of the mTOR downstream effector ribosomal protein S6 as well as other pathway interactors such as Akt and GSK3β. Furthermore, we quantified the status of these proteins in the brain cortex of rats chronically treated with the antipsychotics haloperidol, clozapine, or risperidone. We found a striking decrease in the expression of total S6 and in its active phosphorylated form phospho-S6 (Ser235/236) in the brain of subjects with schizophrenia compared to matched controls. The chronic treatment with the antipsychotics haloperidol and clozapine affected both the expression of GSK3β and the activation of Akt [phospho-Akt (Ser473)] in rat brain cortex, while no changes were observed in S6 and phospho-S6 (Ser235/236) protein expression with any antipsychotic treatment. These findings provide further evidence for the involvement of the mTOR-dependent signaling pathway in schizophrenia and suggest that a hypofunctional S6 may have a role in the etiopathogenesis of this disorder.
Collapse
Affiliation(s)
- Inés Ibarra-Lecue
- Department of Pharmacology, University of the Basque Country UPV/EHU and Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Leioa, Spain
| | - Rebeca Diez-Alarcia
- Department of Pharmacology, University of the Basque Country UPV/EHU and Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Benito Morentin
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Section of Forensic Pathology, Basque Institute of Legal Medicine, Bilbao, Spain
| | - J Javier Meana
- Department of Pharmacology, University of the Basque Country UPV/EHU and Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Luis F Callado
- Department of Pharmacology, University of the Basque Country UPV/EHU and Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Leyre Urigüen
- Department of Pharmacology, University of the Basque Country UPV/EHU and Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| |
Collapse
|
16
|
Aharoni R, Schottlender N, Bar-Lev DD, Eilam R, Sela M, Tsoory M, Arnon R. Cognitive impairment in an animal model of multiple sclerosis and its amelioration by glatiramer acetate. Sci Rep 2019; 9:4140. [PMID: 30858445 PMCID: PMC6412002 DOI: 10.1038/s41598-019-40713-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/21/2019] [Indexed: 01/28/2023] Open
Abstract
The severe motor impairment in the MS animal model experimental autoimmune encephalomyelitis (EAE) obstructs the assessment of cognitive functions. We developed an experimental system that evaluates memory faculties in EAE-affected mice, irrespective of their motor performance, enabling the assessment of cognitive impairments along the disease duration, the associated brain damage, and the consequences of glatiramer acetate (GA) treatment on these manifestations. The delayed-non-matching to sample (DNMS) T-maze task, testing working and long term memory was adapted and utilized. Following the appearance of clinical manifestations task performances of the EAE-untreated mice drastically declined. Cognitive impairments were associated with disease severity, as indicated by a significant correlation between the T-maze performance and the clinical symptoms in EAE-untreated mice. GA-treatment conserved cognitive functions, so that despite their exhibited mild motor impairments, the treated mice performed similarly to naïve controls. The cognitive deficit of EAE-mice coincided with inflammatory and neurodegenerative damage to the frontal cortex and the hippocampus; these damages were alleviated by GA-treatment. These combined findings indicate that in addition to motor impairment, EAE leads to substantial impairment of cognitive functions, starting at the early stages and increasing with disease aggravation. GA-treatment, conserves cognitive capacities and prevents its disease related deterioration.
Collapse
Affiliation(s)
- Rina Aharoni
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 761001, Israel.
| | - Nofar Schottlender
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 761001, Israel
| | - Dekel D Bar-Lev
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 761001, Israel
| | - Raya Eilam
- Department of Veterinary Resources, The Weizmann Institute of Science, Rehovot, 761001, Israel
| | - Michael Sela
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 761001, Israel
| | - Michael Tsoory
- Department of Veterinary Resources, The Weizmann Institute of Science, Rehovot, 761001, Israel
| | - Ruth Arnon
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 761001, Israel.
| |
Collapse
|
17
|
Li M, You M, Li S, Qiu Z, Wang Y. Effects of maternal exposure to nonylphenol on learning and memory in offspring involve inhibition of BDNF-PI3K/Akt signaling. Brain Res Bull 2019; 146:270-278. [PMID: 30660719 DOI: 10.1016/j.brainresbull.2019.01.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 01/10/2019] [Accepted: 01/13/2019] [Indexed: 12/21/2022]
Abstract
Nonylphenol (NP), a global environmental pollutant, has been found to result in impairments of neurodevelopment. However, effects of maternal exposure to NP on learning and memory and the potential mechanisms are largely unexplored. Thus, we treated dams with NP during gestation and lactation to study its effect on learning and memory in offspring. Morris water maze (MWM) task and the electrophysiological recording in the hippocampus were conducted in pups. We also investigated the activation of BDNF-PI3K/Akt signaling and the expression of its target protein PSD-95 in offspring hippocampus, which are curial for the synaptic plasticity and learning and memory. The results showed that maternal exposure to NP led to poor performance in MWM task and especially impairments of long-term potentiation (LTP), although the termination of NP exposure was at the end of lactation. Meanwhile, maternal exposure to NP also decreased the activation of BDNF-PI3K/Akt signaling and the protein level of PSD-95. Taken together, our results support the hypothesis that maternal exposure to NP during gestation and lactation causes damages to learning and memory. In addition, suppressed activation of the BDNF-PI3K/Akt signaling may contribute to these impairments caused by maternal exposure to NP.
Collapse
Affiliation(s)
- Mei Li
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, PR China
| | - Mingdan You
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, PR China
| | - Siyao Li
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, PR China
| | - Zhenmin Qiu
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, PR China
| | - Yi Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, PR China.
| |
Collapse
|
18
|
The effects of donepezil on phencyclidine-induced cognitive deficits in a mouse model of schizophrenia. Pharmacol Biochem Behav 2018; 175:69-76. [PMID: 30218672 DOI: 10.1016/j.pbb.2018.09.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/09/2018] [Accepted: 09/11/2018] [Indexed: 01/19/2023]
Abstract
Donepezil is the first-line of treatment for Alzheimer's disease (AD), which improves cognitive impairment effectively, but whether it has an impact on cognitive impairment in schizophrenia remains unknown. In this study, we evaluated the effects and mechanisms of donepezil on schizophrenia-like cognitive deficits induced by phencyclidine (PCP). The cognitive deficits model of schizophrenia was established by injecting PCP into mice. Risperidone, an atypical antipsychotic drug, served as positive control drug. Three behavioral tests including novel object recognition (NOR) test, Morris Water Maze (MWM) and passive avoidance (PA) test were performed to evaluate the effect of donepezil on PCP-induced cognitive deficits. Furthermore, the content of BDNF and NGF in the hippocampus and cortex of mice was determined using ELISA. Expressions of p-GSK-3β/GSK-3β, p-Akt/Akt, Bcl-2/Bax and Caspase-3 in the hippocampus and cortex were detected by Western blot. Results revealed that donepezil has a protective effect on PCP-induced cognitive dysfunction. Moreover, donepezil can also improve PCP-induced schizophrenia-like cognitive deficits by inhibiting neuronal apoptosis and regulating synaptic plasticity, which was possible through the up-regulation of p-Akt, p-GSK-3β, Bcl-2 and the down-regulation of Bax, Caspase-3. The results indicated that donepezil might exhibit a beneficial effect on the treatment of cognitive dysfunction in schizophrenia.
Collapse
|
19
|
Chow TJ, Tee SF, Loh SY, Yong HS, Abu Bakar AK, Song SL, Tang PY. Identification of AKT1 3'UTR variants in two Indian schizophrenia patients with poor executive functioning. Asian J Psychiatr 2018; 36:17-18. [PMID: 29864676 DOI: 10.1016/j.ajp.2018.05.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 05/21/2018] [Indexed: 11/19/2022]
Affiliation(s)
- Tze Jen Chow
- Department of Mechatronics and Biomedical Engineering, Lee Kong Chian Faculty of Engineering and Science, Universiti Tunku Abdul Rahman, Bandar Sungai Long, Cheras 43000 Kajang, Malaysia
| | - Shiau Foon Tee
- Department of Chemical Engineering, Lee Kong Chian Faculty of Engineering and Science, Universiti Tunku Abdul Rahman, Bandar Sungai Long, Cheras 43000 Kajang, Malaysia
| | - Siew Yim Loh
- Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Hoi Sen Yong
- Institute of Biological Sciences, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | | | - Sze Looi Song
- Institute of Ocean and Earth Sciences, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Pek Yee Tang
- Department of Mechatronics and Biomedical Engineering, Lee Kong Chian Faculty of Engineering and Science, Universiti Tunku Abdul Rahman, Bandar Sungai Long, Cheras 43000 Kajang, Malaysia.
| |
Collapse
|
20
|
Zamarbide M, Oaks AW, Pond HL, Adelman JS, Manzini MC. Loss of the Intellectual Disability and Autism Gene Cc2d1a and Its Homolog Cc2d1b Differentially Affect Spatial Memory, Anxiety, and Hyperactivity. Front Genet 2018; 9:65. [PMID: 29552027 PMCID: PMC5840150 DOI: 10.3389/fgene.2018.00065] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 02/15/2018] [Indexed: 11/13/2022] Open
Abstract
Hundreds of genes are mutated in non-syndromic intellectual disability (ID) and autism spectrum disorder (ASD), with each gene often involved in only a handful of cases. Such heterogeneity can be daunting, but rare recessive loss of function (LOF) mutations can be a good starting point to provide insight into the mechanisms of neurodevelopmental disease. Biallelic LOF mutations in the signaling scaffold CC2D1A cause a rare form of autosomal recessive ID, sometimes associated with ASD and seizures. In parallel, we recently reported that Cc2d1a-deficient mice present with cognitive and social deficits, hyperactivity and anxiety. In Drosophila, loss of the only ortholog of Cc2d1a, lgd, is embryonically lethal, while in vertebrates, Cc2d1a has a homolog Cc2d1b which appears to be compensating, indicating that Cc2d1a and Cc2d1b have a redundant function in humans and mice. Here, we generate an allelic series of Cc2d1a and Cc2d1b LOF to determine the relative role of these genes during behavioral development. We generated Cc2d1b knockout (KO), Cc2d1a/1b double heterozygous and double KO mice, then performed behavioral studies to analyze learning and memory, social interactions, anxiety, and hyperactivity. We found that Cc2d1a and Cc2d1b have partially overlapping roles. Overall, loss of Cc2d1b is less severe than loss of Cc2d1a, only leading to cognitive deficits, while Cc2d1a/1b double heterozygous animals are similar to Cc2d1a-deficient mice. These results will help us better understand the deficits in individuals with CC2D1A mutations, suggesting that recessive CC2D1B mutations and trans-heterozygous CC2D1A and CC2D1B mutations could also contribute to the genetics of ID.
Collapse
Affiliation(s)
- Marta Zamarbide
- GW Institute for Neurosciences, Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Adam W. Oaks
- GW Institute for Neurosciences, Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Heather L. Pond
- GW Institute for Neurosciences, Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Julia S. Adelman
- GW Institute for Neurosciences, Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - M. Chiara Manzini
- GW Institute for Neurosciences, Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
- Autism and Neurodevelopmental Disorders Institute, The George Washington University, Washington, DC, United States
| |
Collapse
|
21
|
PKBγ/AKT3 loss-of-function causes learning and memory deficits and deregulation of AKT/mTORC2 signaling: Relevance for schizophrenia. PLoS One 2017; 12:e0175993. [PMID: 28467426 PMCID: PMC5414975 DOI: 10.1371/journal.pone.0175993] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 04/04/2017] [Indexed: 12/17/2022] Open
Abstract
Psychiatric genetic studies have identified genome-wide significant loci for schizophrenia. The AKT3/1q44 locus is a principal risk region and gene-network analyses identify AKT3 polymorphisms as a constituent of several neurobiological pathways relevant to psychiatric risk; the neurobiological mechanisms remain unknown. AKT3 shows prenatal enrichment during human neocortical development and recurrent copy number variations involving the 1q43-44 locus are associated with cortical malformations and intellectual disability, implicating an essential role in early brain development. Here, we investigated the role of AKT3 as it relates to aspects of learning and memory and behavioral function, relevant to schizophrenia and cognitive disability, utilizing a novel murine model of Akt3 genetic deficiency. Akt3 heterozygous (Akt3-/+) or null mice (Akt3-/-) were assessed in a comprehensive test battery. Brain biochemical studies were conducted to assess the impact of Akt3 deficiency on cortical Akt/mTOR signaling. Akt3-/+ and Akt3-/- mice exhibited selective deficits of temporal order discrimination and spatial memory, tasks critically dependent on intact prefrontal-hippocampal circuitry, but showed normal prepulse inhibition, fear conditioned learning, memory for novel objects and social function. Akt3 loss-of-function, reduced brain size and dramatically impaired cortical Akt Ser473 activation in an allele-dose dependent manner. Such changes were observed in the absence of altered Akt1 or Akt2 protein expression. Concomitant reduction of the mTORC2 complex proteins, Rictor and Sin1 identifies a potential mechanism. Our findings provide novel insight into the neurodevelopmental role of Akt3, identify a non-redundant role for Akt3 in the development of prefrontal cortical-mediated cognitive function and show that Akt3 is potentially the dominant regulator of AKT/mTOR signaling in brain.
Collapse
|
22
|
Blokland GAM, Wallace AK, Hansell NK, Thompson PM, Hickie IB, Montgomery GW, Martin NG, McMahon KL, de Zubicaray GI, Wright MJ. Genome-wide association study of working memory brain activation. Int J Psychophysiol 2017; 115:98-111. [PMID: 27671502 PMCID: PMC5364069 DOI: 10.1016/j.ijpsycho.2016.09.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 08/05/2016] [Accepted: 09/15/2016] [Indexed: 11/30/2022]
Abstract
In a population-based genome-wide association (GWA) study of n-back working memory task-related brain activation, we extracted the average percent BOLD signal change (2-back minus 0-back) from 46 regions-of-interest (ROIs) in functional MRI scans from 863 healthy twins and siblings. ROIs were obtained by creating spheres around group random effects analysis local maxima, and by thresholding a voxel-based heritability map of working memory brain activation at 50%. Quality control for test-retest reliability and heritability of ROI measures yielded 20 reliable (r>0.7) and heritable (h2>20%) ROIs. For GWA analysis, the cohort was divided into a discovery (n=679) and replication (n=97) sample. No variants survived the stringent multiple-testing-corrected genome-wide significance threshold (p<4.5×10-9), or were replicated (p<0.0016), but several genes were identified that are worthy of further investigation. A search of 529,379 genomic markers resulted in discovery of 31 independent single nucleotide polymorphisms (SNPs) associated with BOLD signal change at a discovery level of p<1×10-5. Two SNPs (rs7917410 and rs7672408) were associated at a significance level of p<1×10-7. Only one, most strongly affecting BOLD signal change in the left supramarginal gyrus (R2=5.5%), had multiple SNPs associated at p<1×10-5 in linkage disequilibrium with it, all located in and around the BANK1 gene. BANK1 encodes a B-cell-specific scaffold protein and has been shown to negatively regulate CD40-mediated AKT activation. AKT is part of the dopamine-signaling pathway, suggesting a mechanism for the involvement of BANK1 in the BOLD response to working memory. Variants identified here may be relevant to (the susceptibility to) common disorders affecting brain function.
Collapse
Affiliation(s)
- Gabriëlla A M Blokland
- QIMR Berghofer Medical Research Institute, Royal Brisbane and Women's Hospital, 300 Herston Road, Brisbane, QLD, 4006, Australia; Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, 4072, Australia; School of Psychology, The University of Queensland, St Lucia, QLD, 4072, Australia.
| | - Angus K Wallace
- QIMR Berghofer Medical Research Institute, Royal Brisbane and Women's Hospital, 300 Herston Road, Brisbane, QLD, 4006, Australia
| | - Narelle K Hansell
- QIMR Berghofer Medical Research Institute, Royal Brisbane and Women's Hospital, 300 Herston Road, Brisbane, QLD, 4006, Australia; Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Paul M Thompson
- Imaging Genetics Center, Institute for Neuroimaging and Informatics, Keck School of Medicine, University of Southern California, 2001 North Soto Street - Room 102, Marina del Rey, Los Angeles, CA 90032, United States
| | - Ian B Hickie
- Brain & Mind Research Institute, The University of Sydney, 94 Mallett Street, Camperdown, NSW 2050, Australia
| | - Grant W Montgomery
- QIMR Berghofer Medical Research Institute, Royal Brisbane and Women's Hospital, 300 Herston Road, Brisbane, QLD, 4006, Australia
| | - Nicholas G Martin
- QIMR Berghofer Medical Research Institute, Royal Brisbane and Women's Hospital, 300 Herston Road, Brisbane, QLD, 4006, Australia
| | - Katie L McMahon
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Greig I de Zubicaray
- School of Psychology, The University of Queensland, St Lucia, QLD, 4072, Australia; Faculty of Health and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Margaret J Wright
- QIMR Berghofer Medical Research Institute, Royal Brisbane and Women's Hospital, 300 Herston Road, Brisbane, QLD, 4006, Australia; Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, 4072, Australia; School of Psychology, The University of Queensland, St Lucia, QLD, 4072, Australia; Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| |
Collapse
|
23
|
Bergeron Y, Bureau G, Laurier-Laurin MÉ, Asselin E, Massicotte G, Cyr M. Genetic Deletion of Akt3 Induces an Endophenotype Reminiscent of Psychiatric Manifestations in Mice. Front Mol Neurosci 2017; 10:102. [PMID: 28442992 PMCID: PMC5385361 DOI: 10.3389/fnmol.2017.00102] [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: 01/26/2017] [Accepted: 03/24/2017] [Indexed: 11/21/2022] Open
Abstract
The protein kinase B (PKB/Akt), found in three distinctive isoforms (PKBα/Akt1, PKBβ/Akt2, PKBγ/Akt3), is implicated in a variety of cellular processes such as cell development, growth and survival. Although Akt3 is the most expressed isoform in the brain, its role in cerebral functions is still unclear. In the present study, we investigated the behavioral, electrophysiological and biochemical consequences of Akt3 deletion in mice. Motor abilities, spatial navigation, recognition memory and LTP are intact in the Akt3 knockout (KO) mice. However, the prepulse inhibition, three-chamber social, forced swim, tail suspension, open field, elevated plus maze and light-dark transition tests revealed an endophenotype reminiscent of psychiatric manifestations such as schizophrenia, anxiety and depression. Biochemical investigations revealed that Akt3 deletion was associated with reduced levels of phosphorylated GSK3α/β at serine 21/9 in several brain regions, although Akt1 and Akt2 levels were unaffected. Notably, chronic administration of lithium, a mood stabilizer, restored the decreased phosphorylated GSK3α/β levels and rescued the depressive and anxiety-like behaviors in the Akt3 KO mice. Collectively, our data suggest that Akt3 might be a critical molecule underlying psychiatric-related behaviors in mice.
Collapse
Affiliation(s)
- Yan Bergeron
- Department of Medical Biology, Université du Québec à Trois-RivièresTrois-Rivières, QC, Canada
| | - Geneviève Bureau
- Department of Medical Biology, Université du Québec à Trois-RivièresTrois-Rivières, QC, Canada
| | | | - Eric Asselin
- Department of Medical Biology, Université du Québec à Trois-RivièresTrois-Rivières, QC, Canada
| | - Guy Massicotte
- Department of Medical Biology, Université du Québec à Trois-RivièresTrois-Rivières, QC, Canada
| | - Michel Cyr
- Department of Medical Biology, Université du Québec à Trois-RivièresTrois-Rivières, QC, Canada
| |
Collapse
|
24
|
Kebir O, Chaumette B, Rivollier F, Miozzo F, Lemieux Perreault LP, Barhdadi A, Provost S, Plaze M, Bourgin J, Gaillard R, Mezger V, Dubé MP, Krebs MO. Methylomic changes during conversion to psychosis. Mol Psychiatry 2017; 22:512-518. [PMID: 27113994 PMCID: PMC5378806 DOI: 10.1038/mp.2016.53] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 02/28/2016] [Accepted: 03/02/2016] [Indexed: 12/12/2022]
Abstract
The onset of psychosis is the consequence of complex interactions between genetic vulnerability to psychosis and response to environmental and/or maturational changes. Epigenetics is hypothesized to mediate the interplay between genes and environment leading to the onset of psychosis. We believe we performed the first longitudinal prospective study of genomic DNA methylation during psychotic transition in help-seeking young individuals referred to a specialized outpatient unit for early detection of psychosis and enrolled in a 1-year follow-up. We used Infinium HumanMethylation450 BeadChip array after bisulfite conversion and analyzed longitudinal variations in methylation at 411 947 cytosine-phosphate-guanine (CpG) sites. Conversion to psychosis was associated with specific methylation changes. Changes in DNA methylation were significantly different between converters and non-converters in two regions: one located in 1q21.1 and a cluster of six CpG located in GSTM5 gene promoter. Methylation data were confirmed by pyrosequencing in the same population. The 100 top CpGs associated with conversion to psychosis were subjected to exploratory analyses regarding the related gene networks and their capacity to distinguish between converters and non-converters. Cluster analysis showed that the top CpG sites correctly distinguished between converters and non-converters. In this first study of methylation during conversion to psychosis, we found that alterations preferentially occurred in gene promoters and pathways relevant for psychosis, including oxidative stress regulation, axon guidance and inflammatory pathways. Although independent replications are warranted to reach definitive conclusions, these results already support that longitudinal variations in DNA methylation may reflect the biological mechanisms that precipitate some prodromal individuals into full-blown psychosis, under the influence of environmental factors and maturational processes at adolescence.
Collapse
Affiliation(s)
- O Kebir
- Université Paris Descartes, PRES Université Paris Sorbonne Paris Cité, Centre de Psychiatrie et Neurosciences, UMR S 894, Paris, France,INSERM, Laboratoire de Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences, UMR S 894, Paris, France,CNRS, GDR3557-Institut de Psychiatrie, Paris, France,Faculté de Médecine Paris Descartes, Centre Hospitalier Sainte-Anne, Service Hospitalo-Universitaire, Paris, France
| | - B Chaumette
- Université Paris Descartes, PRES Université Paris Sorbonne Paris Cité, Centre de Psychiatrie et Neurosciences, UMR S 894, Paris, France,INSERM, Laboratoire de Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences, UMR S 894, Paris, France,CNRS, GDR3557-Institut de Psychiatrie, Paris, France,Faculté de Médecine Paris Descartes, Centre Hospitalier Sainte-Anne, Service Hospitalo-Universitaire, Paris, France
| | - F Rivollier
- Université Paris Descartes, PRES Université Paris Sorbonne Paris Cité, Centre de Psychiatrie et Neurosciences, UMR S 894, Paris, France,INSERM, Laboratoire de Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences, UMR S 894, Paris, France,CNRS, GDR3557-Institut de Psychiatrie, Paris, France,Faculté de Médecine Paris Descartes, Centre Hospitalier Sainte-Anne, Service Hospitalo-Universitaire, Paris, France
| | - F Miozzo
- CNRS, UMR7216 Épigénétique et Destin Cellulaire, Paris, France,Université Paris Diderot, Sorbonne Paris Cité, Paris, France,Département Hospitalo-Universitaire DHU PROTECT, Paris, France
| | - L P Lemieux Perreault
- Université de Montréal, Beaulieu-Saucier Pharmacogenomics Center, Montréal Heart Institute, Montréal, QC, Canada
| | - A Barhdadi
- Université de Montréal, Beaulieu-Saucier Pharmacogenomics Center, Montréal Heart Institute, Montréal, QC, Canada
| | - S Provost
- Université de Montréal, Beaulieu-Saucier Pharmacogenomics Center, Montréal Heart Institute, Montréal, QC, Canada
| | - M Plaze
- Université Paris Descartes, PRES Université Paris Sorbonne Paris Cité, Centre de Psychiatrie et Neurosciences, UMR S 894, Paris, France,INSERM, Laboratoire de Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences, UMR S 894, Paris, France,CNRS, GDR3557-Institut de Psychiatrie, Paris, France,Faculté de Médecine Paris Descartes, Centre Hospitalier Sainte-Anne, Service Hospitalo-Universitaire, Paris, France
| | - J Bourgin
- Université Paris Descartes, PRES Université Paris Sorbonne Paris Cité, Centre de Psychiatrie et Neurosciences, UMR S 894, Paris, France,INSERM, Laboratoire de Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences, UMR S 894, Paris, France,CNRS, GDR3557-Institut de Psychiatrie, Paris, France,Faculté de Médecine Paris Descartes, Centre Hospitalier Sainte-Anne, Service Hospitalo-Universitaire, Paris, France
| | | | - R Gaillard
- Université Paris Descartes, PRES Université Paris Sorbonne Paris Cité, Centre de Psychiatrie et Neurosciences, UMR S 894, Paris, France,INSERM, Laboratoire de Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences, UMR S 894, Paris, France,CNRS, GDR3557-Institut de Psychiatrie, Paris, France,Faculté de Médecine Paris Descartes, Centre Hospitalier Sainte-Anne, Service Hospitalo-Universitaire, Paris, France
| | - V Mezger
- CNRS, UMR7216 Épigénétique et Destin Cellulaire, Paris, France,Université Paris Diderot, Sorbonne Paris Cité, Paris, France,Département Hospitalo-Universitaire DHU PROTECT, Paris, France
| | - M-P Dubé
- Université de Montréal, Beaulieu-Saucier Pharmacogenomics Center, Montréal Heart Institute, Montréal, QC, Canada
| | - M-O Krebs
- Université Paris Descartes, PRES Université Paris Sorbonne Paris Cité, Centre de Psychiatrie et Neurosciences, UMR S 894, Paris, France,INSERM, Laboratoire de Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences, UMR S 894, Paris, France,CNRS, GDR3557-Institut de Psychiatrie, Paris, France,Faculté de Médecine Paris Descartes, Centre Hospitalier Sainte-Anne, Service Hospitalo-Universitaire, Paris, France,Laboratoire de Physiopathologie de Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences, Service Hospitalo-Universitaire, INSERM U894—Université Paris Descartes, 2 ter rue d'Alesia, Paris 75014, France. E-mail:
| |
Collapse
|
25
|
Nunes D, Cruz TL, Jespersen SN, Shemesh N. Mapping axonal density and average diameter using non-monotonic time-dependent gradient-echo MRI. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2017; 277:117-130. [PMID: 28282586 DOI: 10.1016/j.jmr.2017.02.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 02/23/2017] [Accepted: 02/25/2017] [Indexed: 06/06/2023]
Abstract
White Matter (WM) microstructures, such as axonal density and average diameter, are crucial to the normal function of the Central Nervous System (CNS) as they are closely related with axonal conduction velocities. Conversely, disruptions of these microstructural features may result in severe neurological deficits, suggesting that their noninvasive mapping could be an important step towards diagnosing and following pathophysiology. Whereas diffusion based MRI methods have been proposed to map these features, they typically entail the application of powerful gradients, which are rarely available in the clinic, or extremely long acquisition schemes to extract information from parameter-intensive models. In this study, we suggest that simple and time-efficient multi-gradient-echo (MGE) MRI can be used to extract the axon density from susceptibility-driven non-monotonic decay in the time-dependent signal. We show, both theoretically and with simulations, that a non-monotonic signal decay will occur for multi-compartmental microstructures - such as axons and extra-axonal spaces, which were here used as a simple model for the microstructure - and that, for axons parallel to the main magnetic field, the axonal density can be extracted. We then experimentally demonstrate in ex-vivo rat spinal cords that its different tracts - characterized by different microstructures - can be clearly contrasted using the MGE-derived maps. When the quantitative results are compared against ground-truth histology, they reflect the axonal fraction (though with a bias, as evident from Bland-Altman analysis). As well, the extra-axonal fraction can be estimated. The results suggest that our model is oversimplified, yet at the same time evidencing a potential and usefulness of the approach to map underlying microstructures using a simple and time-efficient MRI sequence. We further show that a simple general-linear-model can predict the average axonal diameters from the four model parameters, and map these average axonal diameters in the spinal cords. While clearly further modelling and theoretical developments are necessary, we conclude that salient WM microstructural features can be extracted from simple, SNR-efficient multi-gradient echo MRI, and that this paves the way towards easier estimation of WM microstructure in vivo.
Collapse
Affiliation(s)
- Daniel Nunes
- Champalimaud Neuroscience Programme, Champalimaud Centre for the Unknown, Av. Brasilia 1400-038, Lisbon, Portugal
| | - Tomás L Cruz
- Champalimaud Neuroscience Programme, Champalimaud Centre for the Unknown, Av. Brasilia 1400-038, Lisbon, Portugal
| | - Sune N Jespersen
- Center of Functionally Integrative Neuroscience (CFIN) and MINDLab, Clinical Institute, Aarhus University, Aarhus, Denmark; Department of Physics and Astronomy, Aarhus University, Aarhus, Denmark
| | - Noam Shemesh
- Champalimaud Neuroscience Programme, Champalimaud Centre for the Unknown, Av. Brasilia 1400-038, Lisbon, Portugal.
| |
Collapse
|
26
|
Alteration of Neuronal Excitability and Short-Term Synaptic Plasticity in the Prefrontal Cortex of a Mouse Model of Mental Illness. J Neurosci 2017; 37:4158-4180. [PMID: 28283561 DOI: 10.1523/jneurosci.4345-15.2017] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 02/17/2017] [Accepted: 02/22/2017] [Indexed: 01/28/2023] Open
Abstract
Using a genetic mouse model that faithfully recapitulates a DISC1 genetic alteration strongly associated with schizophrenia and other psychiatric disorders, we examined the impact of this mutation within the prefrontal cortex. Although cortical layering, cytoarchitecture, and proteome were found to be largely unaffected, electrophysiological examination of the mPFC revealed both neuronal hyperexcitability and alterations in short-term synaptic plasticity consistent with enhanced neurotransmitter release. Increased excitability of layer II/III pyramidal neurons was accompanied by consistent reductions in voltage-activated potassium currents near the action potential threshold as well as by enhanced recruitment of inputs arising from superficial layers to layer V. We further observed reductions in both the paired-pulse ratios and the enhanced short-term depression of layer V synapses arising from superficial layers consistent with enhanced neurotransmitter release at these synapses. Recordings from layer II/III pyramidal neurons revealed action potential widening that could account for enhanced neurotransmitter release. Significantly, we found that reduced functional expression of the voltage-dependent potassium channel subunit Kv1.1 substantially contributes to both the excitability and short-term plasticity alterations that we observed. The underlying dysregulation of Kv1.1 expression was attributable to cAMP elevations in the PFC secondary to reduced phosphodiesterase 4 activity present in Disc1 deficiency and was rescued by pharmacological blockade of adenylate cyclase. Our results demonstrate a potentially devastating impact of Disc1 deficiency on neural circuit function, partly due to Kv1.1 dysregulation that leads to a dual dysfunction consisting of enhanced neuronal excitability and altered short-term synaptic plasticity.SIGNIFICANCE STATEMENT Schizophrenia is a profoundly disabling psychiatric illness with a devastating impact not only upon the afflicted but also upon their families and the broader society. Although the underlying causes of schizophrenia remain poorly understood, a growing body of studies has identified and strongly implicated various specific risk genes in schizophrenia pathogenesis. Here, using a genetic mouse model, we explored the impact of one of the most highly penetrant schizophrenia risk genes, DISC1, upon the medial prefrontal cortex, the region believed to be most prominently dysfunctional in schizophrenia. We found substantial derangements in both neuronal excitability and short-term synaptic plasticity-parameters that critically govern neural circuit information processing-suggesting that similar changes may critically, and more broadly, underlie the neural computational dysfunction prototypical of schizophrenia.
Collapse
|
27
|
Zhang J, Wei SY, Yuan L, Kong LL, Zhang SX, Wang ZJ, Wu MN, Qi JS. Davunetide improves spatial learning and memory in Alzheimer's disease-associated rats. Physiol Behav 2017; 174:67-73. [PMID: 28257938 DOI: 10.1016/j.physbeh.2017.02.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 02/25/2017] [Accepted: 02/27/2017] [Indexed: 02/07/2023]
Abstract
Memory loss and cognition decline are the main clinical manifestations of Alzheimer's disease (AD). Amyloid β protein (Aβ) aggregated in the brain is one of the key pathological characteristics of AD and responsible for the deficits in learning and memory. It is reported that davunetide, an octapeptide derived from activity-dependent neuroprotective protein (ADNP), inhibited Aβ aggregation and Aβ-induced neurotoxicity. To further characterize the neuroprotective roles of davunetide and its possible mechanism, the present study investigated the effects of davunetide on Aβ1-42-induced impairments in spatial memory, synaptic plasticity and hippocampal AKT level. In Morris water maze (MWM) test, bilateral intrahippocampal injection of Aβ1-42 significantly increased escape latency and decreased target quadrant swimming time of rats, while three weeks of intranasal application of davunetide reversed the Aβ1-42-induced learning deficits and memory loss in a dose-dependent manner. In vivo field potentiation recording showed that Aβ1-42 suppressed long-term potentiation (LTP) of excitatory postsynaptic potential (fEPSP) in the hippocampal CA1 region of rats, while davunetide effectively blocked the suppression of LTP, without affecting paired-pulse facilitation (PPF). Western blotting experiments showed a significant decrease in the level of hippocampal p-AKT (Ser473), not total AKT, in Aβ1-42 only group, which was mostly antagonized by davunetide treatment. These findings demonstrate that davunetide, probably by enhancing PI3K/AKT pathway, plays an important positive role in attenuating Aβ1-42-induced impairments in spatial memory and synaptic plasticity, suggesting that davunetide could be an effective therapeutic candidate for the prevention and treatment of neurodegenerative disease such as AD.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Shu-Yu Wei
- Department of Severe Liver Disease, The 3ird People's Hospital of Taiyuan, Taiyuan 030012, PR China
| | - Li Yuan
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Lin-Lin Kong
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Sheng-Xiao Zhang
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Zhao-Jun Wang
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Mei-Na Wu
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Jin-Shun Qi
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, PR China.
| |
Collapse
|
28
|
Chow TJ, Tee SF, Yong HS, Tang PY. Genetic Association of TCF4 and AKT1 Gene Variants with the Age at Onset of Schizophrenia. Neuropsychobiology 2017; 73:233-40. [PMID: 27305091 DOI: 10.1159/000446285] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 04/19/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND Age at onset (AAO) is a known prognostic indicator for schizophrenia and is hypothesized to correlate with cognition and symptom severity. TCF4 and AKT1 are schizophrenia risk genes involved in cognitive functions. The current study examined the interactive effects of TCF4 and AKT1 variants with gender, family history of psychiatric disorders and ethnicity on the AAO of schizophrenia. METHODS This study consisted of 322 patients with schizophrenia meeting the DSM-IV criteria. Six single nucleotide polymorphisms (SNPs) of TCF4 (rs12966547, rs8766, rs2958182, rs9960767, rs10401120 and rs17512836) and seven AKT1 SNPs (rs2498804, rs3803304, rs2494732, rs3730358, rs1130214, rs2498784 and rs3803300) were genotyped using the TaqMan® SNP genotyping-based assays method. The relationship of AAO with each variant was investigated using analyses of covariance. RESULTS Among the TCF4 variants, rs12966547 (p = 0.024) and rs8766 (p = 0.021) were significantly associated with earlier AAO. We found a lower average AAO in patients with the AA genotype of rs12966547, while the CT genotype of rs8766 was demonstrated to have a protective effect on AAO. For rs8766, there was significant gene × gender interaction (p = 0.012) in influencing AAO. However, these results were not significant after false discovery rate correction. Significant gene × ethnicity interactions were observed to influence AAO (p < 0.05). The Kaplan-Meier curve of the minor AA genotype of rs12966547 displayed a significant trend (p = 0.008) for onset after 19 years of age. Similarly, the minor CC genotype of rs8766 showed a significantly (p = 0.034) lower AAO compared to the TT genotype. CONCLUSION Our analyses suggest that individual risk genotypes may influence the risk of schizophrenia in an age-specific manner.
Collapse
Affiliation(s)
- Tze Jen Chow
- Department of Mechatronics and Biomedical Engineering, Lee Kong Chian Faculty of Engineering and Science, Tunku Abdul Rahman University, Kajang, Malaysia
| | | | | | | |
Collapse
|
29
|
Early postnatal GABAA receptor modulation reverses deficits in neuronal maturation in a conditional neurodevelopmental mouse model of DISC1. Mol Psychiatry 2016; 21:1449-59. [PMID: 26728564 PMCID: PMC4935661 DOI: 10.1038/mp.2015.203] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 10/21/2015] [Accepted: 11/09/2015] [Indexed: 01/28/2023]
Abstract
Exploring drug targets based on disease-associated molecular mechanisms during development is crucial for the generation of novel prevention and treatment strategies for neurodevelopmental psychiatric conditions. We report that prefrontal cortex (PFC)-specific postnatal knockdown of DISC1 via in utero electroporation combined with an inducible knockdown expression system drives deficits in synaptic GABAA function and dendritic development in pyramidal neurons, as well as abnormalities in sensorimotor gating, albeit without profound memory deficits. We show for the first time that DISC1 is specifically involved in regulating cell surface expression of α2 subunit-containing GABAA receptors in immature developing neurons, but not after full maturation. Notably, pharmacological intervention with α2/3 subtype-selective GABAA receptor positive allosteric modulators during the early postnatal period ameliorates dendritic deficits and behavioral abnormalities induced by knockdown of DISC1. These findings highlight a critical role of DISC1-mediated disruption of postnatal GABA signaling in aberrant PFC maturation and function.
Collapse
|
30
|
Liu L, Luo Y, Zhang G, Jin C, Zhou Z, Cheng Z, Yuan G. The mRNA expression of DRD2, PI3KCB, and AKT1 in the blood of acute schizophrenia patients. Psychiatry Res 2016; 243:397-402. [PMID: 27449010 DOI: 10.1016/j.psychres.2016.07.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 06/03/2016] [Accepted: 07/04/2016] [Indexed: 10/21/2022]
Abstract
The phosphoinositide 3 kinase - protein kinase B (PI3K-Akt) signaling pathway plays an important role in the dopamine D2 receptor (DRD2) pathway and in the pathophysiology of schizophrenia. This study measured the mRNA levels of DRD2, PI3KCB, and AKT1 in peripheral blood samples from 24 acute schizophrenia patients and 20 healthy controls using real-time quantitative reverse transcription polymerase chain reaction (real-time qRT-PCR). We found that in the acute schizophrenia patients, the mRNA expression levels of DRD2 and PI3KCB were significantly lower than those in the healthy controls, while the AKT1 mRNA levels were significantly higher than those in the healthy controls. A significant relationship between the mRNA levels of DRD2 and PI3KCB was found only in the controls. In conclusion, the gene expression state of the DRD2-PI3K-AKT signaling cascade differed significantly between acute schizophrenia patients and healthy controls.
Collapse
Affiliation(s)
- Liang Liu
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China.
| | - Yin Luo
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Guofu Zhang
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Chunhui Jin
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Zhenhe Zhou
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Zaohuo Cheng
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China; Wuxi Tongren International Rehabilitation Hospital, Nanjing Medical University, Wuxi, China
| | - Guozhen Yuan
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China; Wuxi Tongren International Rehabilitation Hospital, Nanjing Medical University, Wuxi, China
| |
Collapse
|
31
|
Chang CY, Chen YW, Wang TW, Lai WS. Akting up in the GABA hypothesis of schizophrenia: Akt1 deficiency modulates GABAergic functions and hippocampus-dependent functions. Sci Rep 2016; 6:33095. [PMID: 27615800 PMCID: PMC5018883 DOI: 10.1038/srep33095] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 08/19/2016] [Indexed: 12/29/2022] Open
Abstract
Accumulating evidence implies that both AKT1 and GABAA receptor (GABAAR) subunit genes are involved in schizophrenia pathogenesis. Activated Akt promotes GABAergic neuron differentiation and increases GABAAR expression on the plasma membrane. To elucidate the role of Akt1 in modulating GABAergic functions and schizophrenia-related cognitive deficits, a set of 6 in vitro and in vivo experiments was conducted. First, an Akt1/2 inhibitor was applied to evaluate its effect on GABAergic neuron-like cell formation from P19 cells. Inhibiting Akt resulted in a reduction in parvalbumin-positive neuron-like cells. In Akt1−/− and wild-type mice, seizures induced using pentylenetetrazol (a GABAAR antagonist) were measured, and GABAAR expression and GABAergic interneuron abundance in the brain were examined. Female Akt1−/− mice, but not male Akt1−/− mice, exhibited less pentylenetetrazol-induced convulsive activity than their corresponding wild-type controls. Reduced parvalbumin-positive interneuron abundance and GABAAR subunit expression, especially in the hippocampus, were also observed in female Akt1−/− mice compared to female wild-type mice. Neuromorphometric analyses revealed significantly reduced neurite complexity in hippocampal pyramidal neurons. Additionally, female Akt1−/− mice displayed increased hippocampal oscillation power and impaired spatial memory compared to female wild-type mice. Our findings suggest that Akt1 deficiency modulates GABAergic interneurons and GABAAR expression, contributing to hippocampus-dependent cognitive functional impairment.
Collapse
Affiliation(s)
- Chia-Yuan Chang
- Department of Psychology, National Taiwan University, Taipei, Taiwan
| | - Yi-Wen Chen
- Department of Psychology, National Taiwan University, Taipei, Taiwan
| | - Tsu-Wei Wang
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Wen-Sung Lai
- Department of Psychology, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Brain and Mind Sciences, National Taiwan University, Taipei, Taiwan.,Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
32
|
Huang CW, Hong TW, Wang YJ, Chen KC, Pei JC, Chuang TY, Lai WS, Tsai SH, Chu R, Chen WC, Sheen LY, Takahashi S, Ding ST, Shen TL. Ophiocordyceps formosana improves hyperglycemia and depression-like behavior in an STZ-induced diabetic mouse model. Altern Ther Health Med 2016; 16:310. [PMID: 27553852 PMCID: PMC4995616 DOI: 10.1186/s12906-016-1278-7] [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: 12/25/2015] [Accepted: 08/11/2016] [Indexed: 12/27/2022]
Abstract
Background A newly defined Cordyceps species, Ophiocordyceps formosana (O. formosana) has been implicated in multitudinous bioactivities, including lowering glucose and cholesterol levels and modulating the immune system. However, few literatures demonstrate sufficient evidence to support these proposed functions. Although the use of Cordyceps spp. has been previously addressed to improve insulin insensitivity and improve the detrimental symptoms of depression; its mechanistic nature remains unsettled. Herein, we reveal the effects of O. formosana in ameliorating hyperglycemia accompanied with depression. Methods Diabetes was induced in mice by employing streptozotocin(STZ), a chemical that is toxic to insulin-producing β cells of the pancreas. These streptozotocin (STZ)-induced diabetic mice showed combined symptoms of hyperglycemia and depressive behaviors. Twenty-four STZ-induced mice were randomly divided into 3 groups subjected to oral gavage with 100 μL solution of either PBS or 25 mg/mL Ophiocordyceps formosana extract (OFE) or 2 mg/mL rosiglitazone (Rosi, positive control group). Treatments were administered once per day for 28 days. An additional 6 mice without STZ induction were treated with PBS to serve as the control group. Insulin sensitivity was measured by a glucose tolerance test and levels of adiponectin in plasma and adipose tissue were also quantified. Behavioral tests were conducted and levels of monoamines in various brain regions relating to depression were evaluated. Results HPLC analysis uncovered three major constituents, adenosine, D-mannitol and cordycepin, within O. formosana similar to other prestigious medicinal Cordyceps spp.. STZ-induced diabetic mice demonstrated decreased body weight and subcutaneous adipose tissue, while these symptoms were recovered in mice receiving OFE treatment. Moreover, the OFE group displayed improved insulin sensitivity and elevated adiponectin within the plasma and adipose tissue. The anti-depressive effect of OFE was observed in various depression-related behavior tests. Concurrently, neurotransmitters, like 5-HT and dopamine in the frontal cortex, striatum and hippocampus were found to be up-regulated in OFE-treated mice. Conclusions Our findings illustrated, for the first time, the medicinal merits of O. formosana on Type I diabetes and hyperglycemia-induced depression. OFE were found to promote the expression of adiponectin, which is an adipokine involved in insulin sensitivity and hold anti-depressive effects. In addition, OFE administration also displayed altered levels of neurotransmitters in certain brain regions that may have contributed to its anti-depressive effect. Collectively, this current study provided insights to the potential therapeutic effects of O. formosana extracts in regards to hyperglycemia and its depressive complications.
Collapse
|
33
|
Li YC, Yang SS, Gao WJ. Disruption of Akt signaling decreases dopamine sensitivity in modulation of inhibitory synaptic transmission in rat prefrontal cortex. Neuropharmacology 2016; 108:403-14. [PMID: 27163190 DOI: 10.1016/j.neuropharm.2016.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 04/28/2016] [Accepted: 05/05/2016] [Indexed: 11/18/2022]
Abstract
Akt is a serine/threonine kinase, which is dramatically reduced in the prefrontal cortex (PFC) of patients with schizophrenia, and a deficiency in Akt1 results in PFC function abnormalities. Although the importance of Akt in dopamine (DA) transmission is well established, how impaired Akt signaling affects the DA modulation of synaptic transmission in the PFC has not been characterized. Here we show that Akt inhibitors significantly decreased receptor sensitivity to DA by shifting DA modulation of GABAA receptor-mediated inhibitory postsynaptic currents (IPSCs) in prefrontal cortical neurons. Akt inhibition caused a significant decrease in synaptic dopamine D2 receptor (D2R) levels with high-dose DA exposure. In addition, Akt inhibition failed to affect DA modulation of IPSCs after blockade of β-arrestin 2. β-arrestin 2-mediated interaction of clathrin with D2R was enhanced by co-application of a Akt inhibitor and DA. Taken together, the reduced response in DA modulation of inhibitory transmission mainly involved β-arrestin 2-dependent D2R desensitization.
Collapse
Affiliation(s)
- Yan-Chun Li
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Sha-Sha Yang
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Wen-Jun Gao
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA.
| |
Collapse
|
34
|
Pen Y, Borovok N, Reichenstein M, Sheinin A, Michaelevski I. Membrane-tethered AKT kinase regulates basal synaptic transmission and early phase LTP expression by modulation of post-synaptic AMPA receptor level. Hippocampus 2016; 26:1149-67. [PMID: 27068236 DOI: 10.1002/hipo.22597] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2016] [Indexed: 01/17/2023]
Abstract
The serine/threonine kinase AKT/PKB plays a fundamental role in a wide variety of neuronal functions, including neuronal cell development, axonal growth, and synaptic plasticity. Multiple evidence link AKT signaling pathways to regulation of late phase long-term synaptic plasticity, synaptogenesis, and spinogenesis, as well as long-term memory formation. Nevertheless, the downstream effectors mediating the effects of AKT on early phase long-term potentiation (eLTP) are currently unknown. Here we report that using different regimes of pharmacological activation and inhibition of AKT activity in acute hippocampal slices, we found that AKT regulates the post-synaptic expression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPA) receptors affecting solely the expression of eLTP, with no effect on its induction and maintenance. We further show that both maintenance of basal synaptic activity and expression of eLTP require plasma membrane tethering by activated AKT and that basal synaptic activity may be regulated via the direct effects of AKT1 on the expression level of post-synaptic AMPA receptors bypassing the canonical AKT signaling. Finally, we establish that eLTP expression requires the involvement of both the canonical AKT signaling pathways and the direct effect of AKT1 on AMPA receptor activity/expression in the post-synaptic membrane. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Y Pen
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel-Aviv, Israel
| | - N Borovok
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel-Aviv, Israel
| | - M Reichenstein
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel-Aviv, Israel
| | - A Sheinin
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel-Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel-Aviv, Israel
| | - I Michaelevski
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel-Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel-Aviv, Israel
| |
Collapse
|
35
|
Enriquez-Barreto L, Morales M. The PI3K signaling pathway as a pharmacological target in Autism related disorders and Schizophrenia. MOLECULAR AND CELLULAR THERAPIES 2016; 4:2. [PMID: 26877878 PMCID: PMC4751644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 01/25/2016] [Indexed: 11/21/2023]
Abstract
This review is focused in PI3K's involvement in two widespread mental disorders: Autism and Schizophrenia. A large body of evidence points to synaptic dysfunction as a cause of these diseases, either during the initial phases of brain synaptic circuit's development or later modulating synaptic function and plasticity. Autism related disorders and Schizophrenia are complex genetic conditions in which the identification of gene markers has proved difficult, although the existence of single-gene mutations with a high prevalence in both diseases offers insight into the role of the PI3K signaling pathway. In the brain, components of the PI3K pathway regulate synaptic formation and plasticity; thus, disruption of this pathway leads to synapse dysfunction and pathological behaviors. Here, we recapitulate recent evidences that demonstrate the imbalance of several PI3K elements as leading causes of Autism and Schizophrenia, together with the plausible new pharmacological paths targeting this signaling pathway.
Collapse
Affiliation(s)
- Lilian Enriquez-Barreto
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Miguel Morales
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
36
|
Enriquez-Barreto L, Morales M. The PI3K signaling pathway as a pharmacological target in Autism related disorders and Schizophrenia. MOLECULAR AND CELLULAR THERAPIES 2016; 4:2. [PMID: 26877878 PMCID: PMC4751644 DOI: 10.1186/s40591-016-0047-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 01/25/2016] [Indexed: 01/01/2023]
Abstract
This review is focused in PI3K’s involvement in two widespread mental disorders: Autism and Schizophrenia. A large body of evidence points to synaptic dysfunction as a cause of these diseases, either during the initial phases of brain synaptic circuit’s development or later modulating synaptic function and plasticity. Autism related disorders and Schizophrenia are complex genetic conditions in which the identification of gene markers has proved difficult, although the existence of single-gene mutations with a high prevalence in both diseases offers insight into the role of the PI3K signaling pathway. In the brain, components of the PI3K pathway regulate synaptic formation and plasticity; thus, disruption of this pathway leads to synapse dysfunction and pathological behaviors. Here, we recapitulate recent evidences that demonstrate the imbalance of several PI3K elements as leading causes of Autism and Schizophrenia, together with the plausible new pharmacological paths targeting this signaling pathway.
Collapse
Affiliation(s)
- Lilian Enriquez-Barreto
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Miguel Morales
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
37
|
Landek-Salgado MA, Faust TE, Sawa A. Molecular substrates of schizophrenia: homeostatic signaling to connectivity. Mol Psychiatry 2016; 21:10-28. [PMID: 26390828 PMCID: PMC4684728 DOI: 10.1038/mp.2015.141] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Revised: 06/24/2015] [Accepted: 06/25/2015] [Indexed: 02/06/2023]
Abstract
Schizophrenia (SZ) is a devastating psychiatric condition affecting numerous brain systems. Recent studies have identified genetic factors that confer an increased risk of SZ and participate in the disease etiopathogenesis. In parallel to such bottom-up approaches, other studies have extensively reported biological changes in patients by brain imaging, neurochemical and pharmacological approaches. This review highlights the molecular substrates identified through studies with SZ patients, namely those using top-down approaches, while also referring to the fruitful outcomes of recent genetic studies. We have subclassified the molecular substrates by system, focusing on elements of neurotransmission, targets in white matter-associated connectivity, immune/inflammatory and oxidative stress-related substrates, and molecules in endocrine and metabolic cascades. We further touch on cross-talk among these systems and comment on the utility of animal models in charting the developmental progression and interaction of these substrates. Based on this comprehensive information, we propose a framework for SZ research based on the hypothesis of an imbalance in homeostatic signaling from immune/inflammatory, oxidative stress, endocrine and metabolic cascades that, at least in part, underlies deficits in neural connectivity relevant to SZ. Thus, this review aims to provide information that is translationally useful and complementary to pathogenic hypotheses that have emerged from genetic studies. Based on such advances in SZ research, it is highly expected that we will discover biomarkers that may help in the early intervention, diagnosis or treatment of SZ.
Collapse
Affiliation(s)
- M A Landek-Salgado
- Department of Psychiatry, John Hopkins University School of Medicine, Baltimore, MD, USA
| | - T E Faust
- Department of Psychiatry, John Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Neuroscience, John Hopkins University School of Medicine, Baltimore, MD, USA
| | - A Sawa
- Department of Psychiatry, John Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
38
|
Berry M, Ahmed Z, Morgan-Warren P, Fulton D, Logan A. Prospects for mTOR-mediated functional repair after central nervous system trauma. Neurobiol Dis 2015; 85:99-110. [PMID: 26459109 DOI: 10.1016/j.nbd.2015.10.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/09/2015] [Accepted: 10/08/2015] [Indexed: 02/07/2023] Open
Abstract
Recent research has suggested that the growth of central nervous system (CNS) axons during development is mediated through the PI3K/Akt/mammalian target of rapamycin (mTOR) intracellular signalling axis and that suppression of activity in this pathway occurs during maturity as levels of the phosphatase and tensin homologue (PTEN) rise and inhibit PI3K activation of mTOR, accounting for the failure of axon regeneration in the injured adult CNS. This hypothesis is supported by findings confirming that suppression of PTEN in experimental adult animals promotes impressive axon regeneration in the injured visual and corticospinal motor systems. This review focuses on these recent developments, discussing the therapeutic potential of a mTOR-based treatment aimed at promoting functional recovery in CNS trauma patients, recognising that to fulfil this ambition, the new therapy should aim to promote not only axon regeneration but also remyelination of regenerated axons, neuronal survival and re-innervation of denervated targets through accurate axonal guidance and synaptogenesis, all with minimal adverse effects. The translational challenges presented by the implementation of this new axogenic therapy are also discussed.
Collapse
Affiliation(s)
- Martin Berry
- Neurotrauma Research Group, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Zubair Ahmed
- Neurotrauma Research Group, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| | - Peter Morgan-Warren
- Neurotrauma Research Group, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Daniel Fulton
- Neurotrauma Research Group, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Ann Logan
- Neurotrauma Research Group, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| |
Collapse
|
39
|
Abstract
Objects making up complex porous systems in Nature usually span a range of sizes. These size distributions play fundamental roles in defining the physicochemical, biophysical and physiological properties of a wide variety of systems - ranging from advanced catalytic materials to Central Nervous System diseases. Accurate and noninvasive measurements of size distributions in opaque, three-dimensional objects, have thus remained long-standing and important challenges. Herein we describe how a recently introduced diffusion-based magnetic resonance methodology, Non-Uniform-Oscillating-Gradient-Spin-Echo (NOGSE), can determine such distributions noninvasively. The method relies on its ability to probe confining lengths with a (length)6 parametric sensitivity, in a constant-time, constant-number-of-gradients fashion; combined, these attributes provide sufficient sensitivity for characterizing the underlying distributions in μm-scaled cellular systems. Theoretical derivations and simulations are presented to verify NOGSE's ability to faithfully reconstruct size distributions through suitable modeling of their distribution parameters. Experiments in yeast cell suspensions - where the ground truth can be determined from ancillary microscopy - corroborate these trends experimentally. Finally, by appending to the NOGSE protocol an imaging acquisition, novel MRI maps of cellular size distributions were collected from a mouse brain. The ensuing micro-architectural contrasts successfully delineated distinctive hallmark anatomical sub-structures, in both white matter and gray matter tissues, in a non-invasive manner. Such findings highlight NOGSE's potential for characterizing aberrations in cellular size distributions upon disease, or during normal processes such as development.
Collapse
Affiliation(s)
- Noam Shemesh
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Gonzalo A. Álvarez
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Lucio Frydman
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot, 76100, Israel
- * E-mail:
| |
Collapse
|
40
|
Ichikawa T, Nakahata S, Tamura T, Manachai N, Morishita K. The loss of NDRG2 expression improves depressive behavior through increased phosphorylation of GSK3β. Cell Signal 2015. [PMID: 26208882 DOI: 10.1016/j.cellsig.2015.07.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
N-myc downstream-regulated gene 2 (NDRG2) is one of the important stress-inducible genes and plays a critical role in negatively regulating PI3K/AKT signaling during hypoxia and inflammation. Through recruitment of PP2A phosphatase, NDRG2 maintains the dephosphorylated status of PTEN to suppress excessive PI3K/AKT signaling, and loss of NDRG2 expression is frequently seen in various types of cancer with enhanced activation of PI3K/AKT signaling. Because NDRG2 is highly expressed in the nervous system, we investigated whether NDRG2 plays a functional role in the nervous system using Ndrg2-deficient mice. Ndrg2-deficient mice do not display any gross abnormalities in the nervous system, but they have a diminished behavioral response associated with anxiety. Ndrg2-deficient mice exhibited decreased immobility and increased head-dipping and rearing behavior in two behavioral models, indicating an improvement of emotional anxiety-like behavior. Moreover, treatment of wild-type mice with the antidepressant drug imipramine reduced the expression of Ndrg2 in the frontal cortex, which was due to the degradation of HIF-1α through reduced expression of HSP90 protein. Furthermore, we found that the down-regulation of Ndrg2 in Ndrg2-deficient mice and imipramine treatment improved mood behavior with enhanced phosphorylation of GSK3β through activation of PI3K/AKT signaling, suggesting that the expression level of NDRG2 has a causal influence on mood-related phenotypes. Collectively, these results suggest that NDRG2 may be a potential target for mood disorders such as depression and anxiety.
Collapse
Affiliation(s)
- Tomonaga Ichikawa
- Division of Tumor and Cellular Biochemistry, Department of Medical Sciences, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Shingo Nakahata
- Division of Tumor and Cellular Biochemistry, Department of Medical Sciences, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Tomohiro Tamura
- Division of Tumor and Cellular Biochemistry, Department of Medical Sciences, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Nawin Manachai
- Division of Tumor and Cellular Biochemistry, Department of Medical Sciences, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Kazuhiro Morishita
- Division of Tumor and Cellular Biochemistry, Department of Medical Sciences, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan.
| |
Collapse
|
41
|
A framework for analyzing the relationship between gene expression and morphological, topological, and dynamical patterns in neuronal networks. J Neurosci Methods 2015; 245:1-14. [PMID: 25724320 DOI: 10.1016/j.jneumeth.2015.02.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 02/18/2015] [Indexed: 01/15/2023]
Abstract
BACKGROUND A key point in developmental biology is to understand how gene expression influences the morphological and dynamical patterns that are observed in living beings. NEW METHOD In this work we propose a methodology capable of addressing this problem that is based on estimating the mutual information and Pearson correlation between the intensity of gene expression and measurements of several morphological properties of the cells. A similar approach is applied in order to identify effects of gene expression over the system dynamics. Neuronal networks were artificially grown over a lattice by considering a reference model used to generate artificial neurons. The input parameters of the artificial neurons were determined according to two distinct patterns of gene expression and the dynamical response was assessed by considering the integrate-and-fire model. RESULTS As far as single gene dependence is concerned, we found that the interaction between the gene expression and the network topology, as well as between the former and the dynamics response, is strongly affected by the gene expression pattern. In addition, we observed a high correlation between the gene expression and some topological measurements of the neuronal network for particular patterns of gene expression. COMPARISON WITH EXISTING METHODS To our best understanding, there are no similar analyses to compare with. CONCLUSIONS A proper understanding of gene expression influence requires jointly studying the morphology, topology, and dynamics of neurons. The proposed framework represents a first step towards predicting gene expression patterns from morphology and connectivity.
Collapse
|
42
|
Huang CH, Pei JC, Luo DZ, Chen C, Chen YW, Lai WS. Investigation of gene effects and epistatic interactions between Akt1 and neuregulin 1 in the regulation of behavioral phenotypes and social functions in genetic mouse models of schizophrenia. Front Behav Neurosci 2015; 8:455. [PMID: 25688191 PMCID: PMC4310298 DOI: 10.3389/fnbeh.2014.00455] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Accepted: 12/18/2014] [Indexed: 11/13/2022] Open
Abstract
Accumulating evidence from human genetic studies has suggested several functional candidate genes that might contribute to susceptibility to schizophrenia, including AKT1 and neuregulin 1 (NRG1). Recent findings also revealed that NRG1 stimulates the PI3-kinase/AKT signaling pathway, which might be involved in the functional outcomes of some schizophrenic patients. The aim of this study was to evaluate the effect of Akt1-deficiency and Nrg1-deficiency alone or in combination in the regulation of behavioral phenotypes, cognition, and social functions using genetically modified mice as a model. Male Akt1+/−, Nrg1+/−, and double mutant mice were bred and compared with their wild-type (WT) littermate controls. In Experiment 1, general physical examination revealed that all mutant mice displayed a normal profile of body weight during development and a normal brain activity with microPET scan. In Experiment 2, no significant genotypic differences were found in our basic behavioral phenotyping, including locomotion, anxiety-like behavior, and sensorimotor gating function. However, both Nrg1+/− and double mutant mice exhibited impaired episodic-like memory. Double mutant mice also had impaired sociability. In Experiment 3, a synergistic epistasis between Akt1 and Nrg1 was further confirmed in double mutant mice in that they had impaired social interaction compared to the other 3 groups, especially encountering with a novel male or an ovariectomized female. Double mutant and Nrg1+/− mice also emitted fewer female urine-induced ultrasonic vocalization calls. Collectively, our results indicate that double deficiency of Akt1 and Nrg1 can result in the impairment of social cognitive functions, which might be pertinent to the pathogenesis of schizophrenia-related social cognition.
Collapse
Affiliation(s)
- Ching-Hsun Huang
- Department of Psychology, National Taiwan University Taipei, Taiwan
| | - Ju-Chun Pei
- Department of Psychology, National Taiwan University Taipei, Taiwan
| | - Da-Zhong Luo
- Department of Psychology, National Taiwan University Taipei, Taiwan
| | - Ching Chen
- Department of Psychology, National Taiwan University Taipei, Taiwan
| | - Yi-Wen Chen
- Department of Psychology, National Taiwan University Taipei, Taiwan
| | - Wen-Sung Lai
- Department of Psychology, National Taiwan University Taipei, Taiwan ; Graduate Institute of Brain and Mind Sciences, National Taiwan University Taipei, Taiwan ; Neurobiology and Cognitive Science Center, National Taiwan University Taipei, Taiwan
| |
Collapse
|
43
|
Gao Y, Peterson S, Masri B, Hougland MT, Adham N, Gyertyán I, Kiss B, Caron MG, El-Mallakh RS. Cariprazine exerts antimanic properties and interferes with dopamine D2 receptor β-arrestin interactions. Pharmacol Res Perspect 2014; 3:e00073. [PMID: 25692006 PMCID: PMC4317219 DOI: 10.1002/prp2.73] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 02/11/2014] [Accepted: 02/19/2014] [Indexed: 12/27/2022] Open
Abstract
Activation of dopamine D2 receptors (D2R) modulates G protein/cAMP-dependent signaling and also engages Akt-GSK-3 signaling through D2R/β-arrestin 2 scaffolding of Akt and PP2A. This G protein-independent pathway may be important in mediating the antimanic effects of mood stabilizers and antipsychotics. The mood stabilizer lithium influences behavior and Akt/GSK-3 signaling in mice and many antipsychotics have been shown to more potently antagonize the activity of the β-arrestin-2 pathway relative to the G protein-dependent pathway. Cariprazine, an antipsychotic with potent D3R/D2R partial agonist activity and preferential binding to D3R, was investigated for its effects on the mediators of D2R pathways in vitro and its efficacy in animal models of mania. Effects on G protein-dependent activity were measured via inhibition of isoproterenol-induced cAMP production; effects on D2R/β-arrestin 2 signaling were determined using bioluminescence resonance energy transfer (BRET). Cariprazine was tested in vivo for antimanic-like activity, using the ouabain-induced hyperactivity model in rats. Cariprazine was more potent than aripiprazole in inhibiting isoproterenol-induced cAMP although both compounds showed similar maximum efficacy. In assays of D2R/β-arrestin 2-dependent interactions, cariprazine showed very weak partial agonist activity, unless the levels of receptor kinase were increased; as an antagonist it showed similar potency to haloperidol and ∼fivefold greater potency than aripiprazole. In an animal model of mania, cariprazine showed similar efficacy as lithium in attenuating the effects of ouabain-induced hyperactivity. In summary, the differential effects of cariprazine on D2R G protein and β-arrestin 2 mediators of signal transduction pathways could contribute to its potent antimanic-like activity.
Collapse
Affiliation(s)
- Yonglin Gao
- Mood Disorders Research Program, Department of Psychiatry and Behavioral Sciences, University of Louisville School of Medicine MedCenter One, 501 East Broadway, Suite 340, Louisville, Kentucky, 40202
| | - Sean Peterson
- Mood Disorders Research Program, Department of Psychiatry and Behavioral Sciences, University of Louisville School of Medicine MedCenter One, 501 East Broadway, Suite 340, Louisville, Kentucky, 40202
| | - Bernard Masri
- Mood Disorders Research Program, Department of Psychiatry and Behavioral Sciences, University of Louisville School of Medicine MedCenter One, 501 East Broadway, Suite 340, Louisville, Kentucky, 40202
| | - M Tyler Hougland
- Mood Disorders Research Program, Department of Psychiatry and Behavioral Sciences, University of Louisville School of Medicine MedCenter One, 501 East Broadway, Suite 340, Louisville, Kentucky, 40202
| | - Nika Adham
- Mood Disorders Research Program, Department of Psychiatry and Behavioral Sciences, University of Louisville School of Medicine MedCenter One, 501 East Broadway, Suite 340, Louisville, Kentucky, 40202
| | - Istvan Gyertyán
- Mood Disorders Research Program, Department of Psychiatry and Behavioral Sciences, University of Louisville School of Medicine MedCenter One, 501 East Broadway, Suite 340, Louisville, Kentucky, 40202
| | - Béla Kiss
- Mood Disorders Research Program, Department of Psychiatry and Behavioral Sciences, University of Louisville School of Medicine MedCenter One, 501 East Broadway, Suite 340, Louisville, Kentucky, 40202
| | - Marc G Caron
- Mood Disorders Research Program, Department of Psychiatry and Behavioral Sciences, University of Louisville School of Medicine MedCenter One, 501 East Broadway, Suite 340, Louisville, Kentucky, 40202
| | - Rif S El-Mallakh
- Mood Disorders Research Program, Department of Psychiatry and Behavioral Sciences, University of Louisville School of Medicine MedCenter One, 501 East Broadway, Suite 340, Louisville, Kentucky, 40202
| |
Collapse
|
44
|
Crabtree GW, Gogos JA. Synaptic plasticity, neural circuits, and the emerging role of altered short-term information processing in schizophrenia. Front Synaptic Neurosci 2014; 6:28. [PMID: 25505409 PMCID: PMC4243504 DOI: 10.3389/fnsyn.2014.00028] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 10/22/2014] [Indexed: 01/01/2023] Open
Abstract
Synaptic plasticity alters the strength of information flow between presynaptic and postsynaptic neurons and thus modifies the likelihood that action potentials in a presynaptic neuron will lead to an action potential in a postsynaptic neuron. As such, synaptic plasticity and pathological changes in synaptic plasticity impact the synaptic computation which controls the information flow through the neural microcircuits responsible for the complex information processing necessary to drive adaptive behaviors. As current theories of neuropsychiatric disease suggest that distinct dysfunctions in neural circuit performance may critically underlie the unique symptoms of these diseases, pathological alterations in synaptic plasticity mechanisms may be fundamental to the disease process. Here we consider mechanisms of both short-term and long-term plasticity of synaptic transmission and their possible roles in information processing by neural microcircuits in both health and disease. As paradigms of neuropsychiatric diseases with strongly implicated risk genes, we discuss the findings in schizophrenia and autism and consider the alterations in synaptic plasticity and network function observed in both human studies and genetic mouse models of these diseases. Together these studies have begun to point toward a likely dominant role of short-term synaptic plasticity alterations in schizophrenia while dysfunction in autism spectrum disorders (ASDs) may be due to a combination of both short-term and long-term synaptic plasticity alterations.
Collapse
Affiliation(s)
- Gregg W. Crabtree
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia UniversityNew York, NY, USA
| | - Joseph A. Gogos
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia UniversityNew York, NY, USA
- Department of Neuroscience, College of Physicians and Surgeons, Columbia UniversityNew York, NY, USA
| |
Collapse
|
45
|
Meffre D, Massaad C, Grenier J. Lithium chloride stimulates PLP and MBP expression in oligodendrocytes via Wnt/β-catenin and Akt/CREB pathways. Neuroscience 2014; 284:962-971. [PMID: 25451297 DOI: 10.1016/j.neuroscience.2014.10.064] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 10/27/2014] [Accepted: 10/27/2014] [Indexed: 01/20/2023]
Abstract
Deciphering the molecular pathways involved in myelin gene expression is a major point of interest to better understand re/myelination processes. In this study, we investigated the role of Lithium Chloride (LiCl), a drug largely used for the treatment of neurological disorders, on the two major central myelin gene expression (PLP and MBP) in mouse oligodendrocytes. We show that LiCl enhances the expression of both PLP and MBP, by increasing mRNA amount and promoter activities. We investigated whether Wnt/β-catenin and/or Akt/CREB pathways are modulated by LiCl to regulate myelin gene expression. We showed that β-catenin is required both for PLP and MBP basal promoter activities and for LiCl-induced myelin gene stimulation. Furthermore, while CREB functionality does not influence PLP expression, MBP promoter activity depends on Akt/CREB activation. Finally, we show that LiCl can stimulate oligodendrocyte morphological maturation, and promote remyelination after lysolecithin-induced demyelination of organotypic cerebellar slice cultures. Our data provide mechanistic evidences that Akt/CREB together with β-catenin participate in the transcriptional control of PLP and MBP exerted by LiCl. Therefore, the use of LiCl to balance between β-catenin and CREB effectors could be considered as an efficient remyelinating strategy.
Collapse
Affiliation(s)
- D Meffre
- Paris Descartes University, INSERM UMR-S 1124, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France.
| | - C Massaad
- Paris Descartes University, INSERM UMR-S 1124, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - J Grenier
- Paris Descartes University, INSERM UMR-S 1124, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| |
Collapse
|
46
|
Bhattacharyya S, Iyegbe C, Atakan Z, Martin-Santos R, Crippa JA, Xu X, Williams S, Brammer M, Rubia K, Prata D, Collier DA, McGuire PK. Protein kinase B (AKT1) genotype mediates sensitivity to cannabis-induced impairments in psychomotor control. Psychol Med 2014; 44:3315-3328. [PMID: 25065544 DOI: 10.1017/s0033291714000920] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND What determines inter-individual variability to impairments in behavioural control that may underlie road-traffic accidents, and impulsive and violent behaviours occurring under the influence of cannabis, the most widely used illicit drug worldwide? METHOD Employing a double-blind, repeated-measures design, we investigated the genetic and neural basis of variable sensitivity to cannabis-induced behavioural dyscontrol in healthy occasional cannabis users. Acute oral challenge with placebo or Δ9-tetrahydrocannabinol (THC), the main psychoactive ingredient in cannabis, was combined with functional magnetic resonance imaging, while participants performed a response inhibition task that involved inhibiting a pre-potent motor response. They were genotyped for rs1130233 single nucleotide polymorphisms (SNPs) of the protein kinase B (AKT1) gene. RESULTS Errors of inhibition were significantly (p = 0.008) increased following administration of THC in carriers of the A allele, but not in G allele homozygotes of the AKT1 rs1130233 SNP. The A allele carriers also displayed attenuation of left inferior frontal response with THC evident in the sample as a whole, while there was a modest enhancement of inferior frontal activation in the G homozygotes. There was a direct relationship (r = -0.327, p = 0.045) between the behavioural effect of THC and its physiological effect in the inferior frontal gyrus, where AKT1 genotype modulated the effect of THC. CONCLUSIONS These results require independent replication and show that differing vulnerability to acute psychomotor impairments induced by cannabis depends on variation in a gene that influences dopamine function, and is mediated through modulation of the effect of cannabis on the inferior frontal cortex, that is rich in dopaminergic innervation and critical for psychomotor control.
Collapse
Affiliation(s)
- S Bhattacharyya
- Department of Psychosis Studies,King's College London,Institute of Psychiatry, De Crespigny Park, London,UK
| | - C Iyegbe
- Social, Genetic and Developmental Psychiatry Centre,King's College London,Institute of Psychiatry, De Crespigny Park, London,UK
| | - Z Atakan
- Department of Psychosis Studies,King's College London,Institute of Psychiatry, De Crespigny Park, London,UK
| | - R Martin-Santos
- Pharmacology Research Unit, IMIM-Hospital del Mar and Psychiatric Department,ICN,Hospital Clinico, Barcelona,Spain
| | - J A Crippa
- Department of Neurology, Psychiatry and Medical Psychology, Faculty of Medicine of Ribeirão Preto,University of São Paulo,Brazil
| | - X Xu
- Social, Genetic and Developmental Psychiatry Centre,King's College London,Institute of Psychiatry, De Crespigny Park, London,UK
| | - S Williams
- Department of Neuroimaging, Centre for Neuroimaging Sciences,King's College London,Institute of Psychiatry, De Crespigny Park, London,UK
| | - M Brammer
- Department of Neuroimaging, Centre for Neuroimaging Sciences,King's College London,Institute of Psychiatry, De Crespigny Park, London,UK
| | - K Rubia
- Department of Child and Adolescent Psychiatry,King's College London,Institute of Psychiatry, De Crespigny Park, London,UK
| | - D Prata
- Department of Psychosis Studies,King's College London,Institute of Psychiatry, De Crespigny Park, London,UK
| | - D A Collier
- Social, Genetic and Developmental Psychiatry Centre,King's College London,Institute of Psychiatry, De Crespigny Park, London,UK
| | - P K McGuire
- Department of Psychosis Studies,King's College London,Institute of Psychiatry, De Crespigny Park, London,UK
| |
Collapse
|
47
|
Poirier GL, Imamura N, Zanoletti O, Sandi C. Social deficits induced by peripubertal stress in rats are reversed by resveratrol. J Psychiatr Res 2014; 57:157-64. [PMID: 24974003 DOI: 10.1016/j.jpsychires.2014.05.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 04/15/2014] [Accepted: 05/28/2014] [Indexed: 11/30/2022]
Abstract
Adolescence is increasingly recognized as a critical period for the development of the social system, through the maturation of social competences and of their underlying neural circuitries. The present study sought to test the utility of resveratrol, a dietary phenol recently reported to have mood lifting properties, in modulating social interaction that is deficient following early life adversity. The main aims were to 1) pharmacologically restore normative social investigation levels dampened by peripubertal stress in rats and 2) identify neural pathways engaged by this pharmacological approach. Following peripubertal (P28-42) stress consisting of unpredictable exposures to fearful experiences, at adulthood the subjects' propensity for social exploration was examined in the three-chamber apparatus, comparing time invested in social or non-social investigation. Administered intraperitoneally 30 min before testing, resveratrol (20 mg/kg) normalized the peripubertal stress-induced social investigation deficit seen in the vehicle group, selectively altering juvenile but not object exploration. Examination of prefrontal cortex subregion protein samples following acute resveratrol treatment in a separate cohort revealed that while monoamine oxidase A (MAOA) enzymatic activity remained unaltered, nuclear AKT activation was selectively increased in the infralimbic cortex, but not in the prelimbic or anterior cingulate cortex. In contrast, androgen receptor nuclear localization was increased in the prelimbic cortex, but not in the infralimbic or anterior cingulate cortex. This demonstration that social contact deficits are reversed by resveratrol administration emphasizes a prosocial role for this dietary phenol, and evokes the possibility of developing new treatments for social dysfunctions.
Collapse
Affiliation(s)
- Guillaume L Poirier
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| | - Natsuko Imamura
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Olivia Zanoletti
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
48
|
de Bartolomeis A, Buonaguro EF, Iasevoli F, Tomasetti C. The emerging role of dopamine-glutamate interaction and of the postsynaptic density in bipolar disorder pathophysiology: Implications for treatment. J Psychopharmacol 2014; 28:505-26. [PMID: 24554693 DOI: 10.1177/0269881114523864] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aberrant synaptic plasticity, originating from abnormalities in dopamine and/or glutamate transduction pathways, may contribute to the complex clinical manifestations of bipolar disorder (BD). Dopamine and glutamate systems cross-talk at multiple levels, such as at the postsynaptic density (PSD). The PSD is a structural and functional protein mesh implicated in dopamine and glutamate-mediated synaptic plasticity. Proteins at PSD have been demonstrated to be involved in mood disorders pathophysiology and to be modulated by antipsychotics and mood stabilizers. On the other side, post-receptor effectors such as protein kinase B (Akt), glycogen synthase kinase-3 (GSK-3) and the extracellular signal-regulated kinase (Erk), which are implicated in both molecular abnormalities and treatment of BD, may interact with PSD proteins, and participate in the interplay of the dopamine-glutamate signalling pathway. In this review, we describe emerging evidence on the molecular cross-talk between dopamine and glutamate signalling in BD pathophysiology and pharmacological treatment, mainly focusing on dysfunctions in PSD molecules. We also aim to discuss future therapeutic strategies that could selectively target the PSD-mediated signalling cascade at the crossroads of dopamine-glutamate neurotransmission.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Section of Psychiatry, University Medical School of Naples "Federico II", Naples, Italy
| | - Elisabetta F Buonaguro
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Section of Psychiatry, University Medical School of Naples "Federico II", Naples, Italy
| | - Felice Iasevoli
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Section of Psychiatry, University Medical School of Naples "Federico II", Naples, Italy
| | - Carmine Tomasetti
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Section of Psychiatry, University Medical School of Naples "Federico II", Naples, Italy
| |
Collapse
|
49
|
Pei JC, Liu CM, Lai WS. Distinct phenotypes of new transmembrane-domain neuregulin 1 mutant mice and the rescue effects of valproate on the observed schizophrenia-related cognitive deficits. Front Behav Neurosci 2014; 8:126. [PMID: 24782733 PMCID: PMC3995064 DOI: 10.3389/fnbeh.2014.00126] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 03/26/2014] [Indexed: 11/13/2022] Open
Abstract
Accumulating evidence suggests that neuregulin 1 (NRG1) might be involved in the neurodevelopment, neural plasticity, GABAergic neurotransmission, and pathogenesis of schizophrenia. NRG1 is abundantly expressed in the hippocampus, and emerging studies have begun to reveal the link between NRG1 signaling and cognitive deficits in schizophrenic patients. Because the transmembrane domain of NRG1 is vital for both forward and reverse signaling cascades, new Nrg1-deficient mice that carry a truncation of the transmembrane domain of the Nrg1 gene were characterized and used in this study to test a NRG1 loss-of-function hypothesis for schizophrenia. Both male and female Nrg1 heterozygous mutant mice and their wild-type littermates were used in a series of 4 experiments to characterize the impact of Nrg1 on behavioral phenotypes and to determine the importance of Nrg1 in the regulation of hippocampal neuromorphology and local GABAergic interneurons. First, a comprehensive battery of behavioral tasks indicated that male Nrg1-deficient mice exhibited significant impairments in cognitive functions. Second, pharmacological challenges were conducted and revealed that Nrg1 haploinsufficiency altered GABAergic activity in males. Third, although no genotype-specific neuromorphological alterations were found in the hippocampal CA1 pyramidal neurons, significant reductions in the hippocampal expressions of GAD67 and parvalbumin were revealed in the Nrg1-deficient males. Fourth, chronic treatment with valproate rescued the observed behavioral deficits and hippocampal GAD67 reduction in Nrg1-deficient males. Collectively, these results indicate the potential therapeutic effect of valproate and the importance of Nrg1 in the regulation of cognitive functions and hippocampal GABAergic interneurons, especially in males.
Collapse
Affiliation(s)
- Ju-Chun Pei
- Laboratory of Integrated Neuroscience and Ethology, Department of Psychology, National Taiwan University Taipei, Taiwan
| | - Chih-Min Liu
- Department of Psychiatry, National Taiwan University Hospital Taipei, Taiwan
| | - Wen-Sung Lai
- Laboratory of Integrated Neuroscience and Ethology, Department of Psychology, National Taiwan University Taipei, Taiwan ; Graduate Institute of Brain and Mind Sciences, National Taiwan University Taipei, Taiwan ; Neurobiology and Cognitive Science Center, National Taiwan University Taipei, Taiwan
| |
Collapse
|
50
|
Marlinge E, Bellivier F, Houenou J. White matter alterations in bipolar disorder: potential for drug discovery and development. Bipolar Disord 2014; 16:97-112. [PMID: 24571279 DOI: 10.1111/bdi.12135] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Accepted: 05/24/2013] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Brain white matter (WM) alterations have recently emerged as potentially relevant in bipolar disorder. New techniques such as diffusion tensor imaging allow precise exploration of these WM microstructural alterations in bipolar disorder. Our objective was to critically review WM alterations in bipolar disorder, using neuroimaging and neuropathological studies, in the context of neural models and the potential for drug discovery and development. METHODS We conducted a systematic PubMed and Google Scholar search of the WM and bipolar disorder literature up to and including January 2013. RESULTS Findings relating to WM alterations are consistent in neuroimaging and neuropathology studies of bipolar disorder, especially in regions involved in emotional processing such as the anterior frontal lobe, corpus callosum, cingulate cortex, and in fronto-limbic connections. Some of the structural alterations are related to genetic risk factors for bipolar disorder and may underlie the dysfunctional emotional processing described in recent neurobiological models of bipolar disorder. Medication effects in bipolar disorder, from lithium and other mood stabilizers, might impact myelinating processes, particularly by inhibition of glycogen synthase kinase-3 beta. CONCLUSIONS Pathways leading to WM alterations in bipolar disorder represent potential targets for the development and discovery of new drugs. Myelin damage in bipolar disorder suggests that the effects of existing pro-myelinating drugs should also be evaluated to improve our understanding and treatment of this disease.
Collapse
Affiliation(s)
- Emeline Marlinge
- AP-HP, Groupe Henri Mondor-Albert Chenevier, Pôle de Psychiatrie, Paris, France; Inserm, U955, Equipe 15 (Psychiatrie Génétique), Paris, France; Fondation Fondamental, Créteil, France; Neurospin, I2BM, CEA, Gif-Sur-Yvette, France
| | | | | |
Collapse
|