1
|
Maitre M, Taleb O, Jeltsch-David H, Klein C, Mensah-Nyagan AG. Xanthurenic acid: A role in brain intercellular signaling. J Neurochem 2024; 168:2303-2315. [PMID: 38481090 DOI: 10.1111/jnc.16099] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 10/04/2024]
Abstract
Xanthurenic acid (XA) raises a growing multidisciplinary interest based upon its oxidizing properties, its ability to complex certain metal ions, and its detoxifier capacity of 3-hydroxykynurenine (3-HK), its brain precursor. However, little is still known about the role and mechanisms of action of XA in the central nervous system (CNS). Therefore, many research groups have recently investigated XA and its central functions extensively. The present paper critically reviews and discusses all major data related to XA properties and neuronal activities to contribute to the improvement of the current knowledge on XA's central roles and mechanisms of action. In particular, our data showed the existence of a specific G-protein-coupled receptor (GPCR) for XA localized exclusively in brain neurons exhibiting Ca2+-dependent dendritic release and specific electrophysiological responses. XA properties and central activities suggest a role for this compound in brain intercellular signaling. Indeed, XA stimulates cerebral dopamine (DA) release contrary to its structural analog, kynurenic acid (KYNA). Thus, KYNA/XA ratio could be fundamental in the regulation of brain glutamate and DA release. Cerebral XA may also represent an homeostatic signal between the periphery and several brain regions where XA accumulates easily after peripheral administration. Therefore, XA status in certain psychoses or neurodegenerative diseases seems to be reinforced by its brain-specific properties in balance with its formation and peripheral inputs.
Collapse
Affiliation(s)
- Michel Maitre
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, Strasbourg, France
| | - Omar Taleb
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, Strasbourg, France
| | - Hélène Jeltsch-David
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, Strasbourg, France
- Biotechnologie et signalisation cellulaire, UMR 7242 CNRS/Université de Strasbourg, Illkirch Cedex, France
| | - Christian Klein
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, Strasbourg, France
| | - Ayikoe-Guy Mensah-Nyagan
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, Strasbourg, France
| |
Collapse
|
2
|
Wang X, Ma J, Dong Y, Ren X, Li R, Yang G, She G, Tan Y, Chen S. Exploration on the potential efficacy and mechanism of methyl salicylate glycosides in the treatment of schizophrenia based on bioinformatics, molecular docking and dynamics simulation. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2024; 10:64. [PMID: 39019913 PMCID: PMC11255270 DOI: 10.1038/s41537-024-00484-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 07/02/2024] [Indexed: 07/19/2024]
Abstract
The etiological and therapeutic complexities of schizophrenia (SCZ) persist, prompting exploration of anti-inflammatory therapy as a potential treatment approach. Methyl salicylate glycosides (MSGs), possessing a structural parent nucleus akin to aspirin, are being investigated for their therapeutic potential in schizophrenia. Utilizing bioinformation mining, network pharmacology, molecular docking and dynamics simulation, the potential value and mechanism of MSGs (including MSTG-A, MSTG-B, and Gaultherin) in the treatment of SCZ, as well as the underlying pathogenesis of the disorder, were examined. 581 differentially expressed genes related to SCZ were identified in patients and healthy individuals, with 349 up-regulated genes and 232 down-regulated genes. 29 core targets were characterized by protein-protein interaction (PPI) network, with the top 10 core targets being BDNF, VEGFA, PVALB, KCNA1, GRIN2A, ATP2B2, KCNA2, APOE, PPARGC1A and SCN1A. The pathogenesis of SCZ primarily involves cAMP signaling, neurodegenerative diseases and other pathways, as well as regulation of ion transmembrane transport. Molecular docking analysis revealed that the three candidates exhibited binding activity with certain targets with binding affinities ranging from -4.7 to -109.2 kcal/mol. MSTG-A, MSTG-B and Gaultherin show promise for use in the treatment of SCZ, potentially through their ability to modulate the expression of multiple genes involved in synaptic structure and function, ion transport, energy metabolism. Molecular dynamics simulation revealed good binding abilities between MSTG-A, MSTG-B, Gaultherin and ATP2B2. It suggests new avenues for further investigation in this area.
Collapse
Affiliation(s)
- Xiuhuan Wang
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, 100096, PR China
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, PR China
| | - Jiamu Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, PR China
| | - Ying Dong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, PR China
| | - Xueyang Ren
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, PR China
| | - Ruoming Li
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, 100096, PR China
| | - Guigang Yang
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, 100096, PR China
| | - Gaimei She
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, PR China.
| | - Yunlong Tan
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, 100096, PR China.
| | - Song Chen
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, 100096, PR China.
| |
Collapse
|
3
|
Yang KC, Yang BH, Liu MN, Liou YJ, Chou YH. Cognitive impairment in schizophrenia is associated with prefrontal-striatal functional hypoconnectivity and striatal dopaminergic abnormalities. J Psychopharmacol 2024; 38:515-525. [PMID: 38853592 DOI: 10.1177/02698811241257877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
BACKGROUND A better understanding of the mechanisms underlying cognitive impairment in schizophrenia is imperative, as it causes poor functional outcomes and a lack of effective treatments. AIMS This study aimed to investigate the relationships of two proposed main pathophysiology of schizophrenia, altered prefrontal-striatal connectivity and the dopamine system, with cognitive impairment and their interactions. METHODS Thirty-three patients with schizophrenia and 27 healthy controls (HCs) who are right-handed and matched for age and sex were recruited. We evaluated their cognition, functional connectivity (FC) between the dorsolateral prefrontal cortex (DLPFC)/middle frontal gyrus (MiFG) and striatum, and the availability of striatal dopamine transporter (DAT) using a cognitive battery investigating attention, memory, and executive function, resting-state functional magnetic resonance imaging with group independent component analysis and single-photon emission computed tomography with 99mTc-TRODAT. RESULTS Patients with schizophrenia exhibited poorer cognitive performance, reduced FC between DLPFC/MiFG and the caudate nucleus (CN) or putamen, decreased DAT availability in the left CN, and decreased right-left DAT asymmetry in the CN compared to HCs. In patients with schizophrenia, altered imaging markers are associated with cognitive impairments, especially the relationship between DLPFC/MiFG-putamen FC and attention and between DAT asymmetry in the CN and executive function. CONCLUSIONS This study is the first to demonstrate how prefrontal-striatal hypoconnectivity and altered striatal DAT markers are associated with different domains of cognitive impairment in schizophrenia. More research is needed to evaluate their complex relationships and potential therapeutic implications.
Collapse
Affiliation(s)
- Kai-Chun Yang
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Bang-Hung Yang
- Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Mu-N Liu
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ying-Jay Liou
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yuan-Hwa Chou
- Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Psychiatry, Taichung Veterans General Hospital, Taichung, Taiwan
- The Human Brain Research Center, Taichung Veterans General Hospital, Taichung, Taiwan
| |
Collapse
|
4
|
Nesbit MO, Ahn S, Zou H, Floresco SB, Phillips AG. Potentiation of prefrontal cortex dopamine function by the novel cognitive enhancer d-govadine. Neuropharmacology 2024; 246:109849. [PMID: 38244888 DOI: 10.1016/j.neuropharm.2024.109849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/13/2023] [Accepted: 01/16/2024] [Indexed: 01/22/2024]
Abstract
Cognitive impairment is a debilitating feature of psychiatric disorders including schizophrenia, mood disorders and substance use disorders for which there is a substantial lack of effective therapies. d-Govadine (d-GOV) is a tetrahydroprotoberberine recently shown to significantly enhance working memory and behavioural flexibility in several prefrontal cortex (PFC)-dependent rodent tasks. d-GOV potentiates dopamine (DA) efflux in the mPFC and not the nucleus accumbens, a unique pharmacology that sets it apart from many dopaminergic drugs and likely contributes to its effects on cognitive function. However, specific mechanisms involved in the preferential effects of d-GOV on mPFC DA function remain to be determined. The present study employs brain dialysis in male rats to deliver d-GOV into the mPFC or ventral tegmental area (VTA), while simultaneously sampling DA and norepinephrine (NE) efflux in the mPFC. Intra-PFC delivery or systemic administration of d-GOV preferentially potentiated medial prefrontal DA vs NE efflux. This differential effect of d-GOV on the primary catecholamines known to affect mPFC function further underscores its specificity for the mPFC DA system. Importantly, the potentiating effect of d-GOV on mPFC DA was disrupted when glutamatergic transmission was blocked in either the mPFC or the VTA. We hypothesize that d-GOV acts in the mPFC to engage the mesocortical feedback loop through which prefrontal glutamatergic projections activate a population of VTA DA neurons that specifically project back to the PFC. The activation of a PFC-VTA feedback loop to elevate PFC DA efflux without affecting mesolimbic DA release represents a novel approach to developing pro-cognitive drugs.
Collapse
Affiliation(s)
- Maya O Nesbit
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Soyon Ahn
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Haiyan Zou
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Stan B Floresco
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Anthony G Phillips
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
5
|
Dong B, Yue Y, Dong H, Wang Y. N-methyl-D-aspartate receptor hypofunction as a potential contributor to the progression and manifestation of many neurological disorders. Front Mol Neurosci 2023; 16:1174738. [PMID: 37396784 PMCID: PMC10308130 DOI: 10.3389/fnmol.2023.1174738] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/26/2023] [Indexed: 07/04/2023] Open
Abstract
N-methyl-D-aspartate receptors (NMDA) are glutamate-gated ion channels critical for synaptic transmission and plasticity. A slight variation of NMDAR expression and function can result in devastating consequences, and both hyperactivation and hypoactivation of NMDARs are detrimental to neural function. Compared to NMDAR hyperfunction, NMDAR hypofunction is widely implicated in many neurological disorders, such as intellectual disability, autism, schizophrenia, and age-related cognitive decline. Additionally, NMDAR hypofunction is associated with the progression and manifestation of these diseases. Here, we review the underlying mechanisms of NMDAR hypofunction in the progression of these neurological disorders and highlight that targeting NMDAR hypofunction is a promising therapeutic intervention in some neurological disorders.
Collapse
Affiliation(s)
- Bin Dong
- Department of Geriatrics, Jilin Geriatrics Clinical Research Center, The First Hospital of Jilin University, Changchun, China
| | - Yang Yue
- School of Psychology, Northeast Normal University, Changchun, China
| | - Han Dong
- Department of Geriatrics, Jilin Geriatrics Clinical Research Center, The First Hospital of Jilin University, Changchun, China
| | - Yuehui Wang
- Department of Geriatrics, Jilin Geriatrics Clinical Research Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
6
|
Petanjek Z, Banovac I, Sedmak D, Hladnik A. Dendritic Spines: Synaptogenesis and Synaptic Pruning for the Developmental Organization of Brain Circuits. ADVANCES IN NEUROBIOLOGY 2023; 34:143-221. [PMID: 37962796 DOI: 10.1007/978-3-031-36159-3_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Synaptic overproduction and elimination is a regular developmental event in the mammalian brain. In the cerebral cortex, synaptic overproduction is almost exclusively correlated with glutamatergic synapses located on dendritic spines. Therefore, analysis of changes in spine density on different parts of the dendritic tree in identified classes of principal neurons could provide insight into developmental reorganization of specific microcircuits.The activity-dependent stabilization and selective elimination of the initially overproduced synapses is a major mechanism for generating diversity of neural connections beyond their genetic determination. The largest number of overproduced synapses was found in the monkey and human cerebral cortex. The highest (exceeding adult values by two- to threefold) and most protracted overproduction (up to third decade of life) was described for associative layer IIIC pyramidal neurons in the human dorsolateral prefrontal cortex.Therefore, the highest proportion and extraordinarily extended phase of synaptic spine overproduction is a hallmark of neural circuitry in human higher-order associative areas. This indicates that microcircuits processing the most complex human cognitive functions have the highest level of developmental plasticity. This finding is the backbone for understanding the effect of environmental impact on the development of the most complex, human-specific cognitive and emotional capacities, and on the late onset of human-specific neuropsychiatric disorders, such as autism and schizophrenia.
Collapse
Affiliation(s)
- Zdravko Petanjek
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia.
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia.
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia.
| | - Ivan Banovac
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Dora Sedmak
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ana Hladnik
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
7
|
Khan AQ, Thielen L, Le Pen G, Krebs MO, Kebir O, Groh A, Deest M, Bleich S, Frieling H, Jahn K. Methylation pattern and mRNA expression of synapse-relevant genes in the MAM model of schizophrenia in the time-course of adolescence. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2022; 8:110. [PMID: 36481661 PMCID: PMC9732294 DOI: 10.1038/s41537-022-00319-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 11/16/2022] [Indexed: 12/13/2022]
Abstract
Schizophrenia is highly heritable and aggregating in families, but genetics alone does not exclusively explain the pathogenesis. Many risk factors, including childhood trauma, viral infections, migration, and the use of cannabis, are associated with schizophrenia. Adolescence seems to be the critical period where symptoms of the disease manifest. This work focuses on studying an epigenetic regulatory mechanism (the role of DNA methylation) and its interaction with mRNA expression during development, with a particular emphasis on adolescence. The presumptions regarding the role of aberrant neurodevelopment in schizophrenia were tested in the Methyl-Azoxy-Methanol (MAM) animal model. MAM treatment induces neurodevelopmental disruptions and behavioral deficits in off-springs of the treated animals reminiscent of those observed in schizophrenia and is thus considered a promising model for studying this pathology. On a gestational day-17, adult pregnant rats were treated with the antimitotic agent MAM. Experimental animals were divided into groups and subgroups according to substance treatment (MAM and vehicle agent [Sham]) and age of analysis (pre-adolescent and post-adolescent). Methylation and mRNA expression analysis of four candidate genes, which are often implicated in schizophrenia, with special emphasis on the Dopamine hypothesis i.e., Dopamine receptor D2 (Drd2), and the "co-factors" Disrupted in schizophrenia 1 (DISC1), Synaptophysin (Syp), and Dystrobrevin-binding protein 1 (Dtnbp1), was performed in the Gyrus cingulum (CING) and prefrontal cortex (PFC). Data were analyzed to observe the effect of substance treatment between groups and the impact of adolescence within-group. We found reduced pre-adolescent expression levels of Drd2 in both brain areas under the application of MAM. The "co-factor genes" did not show high deviations in mRNA expression levels but high alterations of methylation rates under the application of MAM (up to ~20%), which diminished in the further time course, reaching a comparable level like in Sham control animals after adolescence. The pre-adolescent reduction in DRD2 expression might be interpreted as downregulation of the receptor due to hyperdopaminergic signaling from the ventral tegmental area (VTA), eventually even to both investigated brain regions. The notable alterations of methylation rates in the three analyzed co-factor genes might be interpreted as attempt to compensate for the altered dopaminergic neurotransmission.
Collapse
Affiliation(s)
- Abdul Qayyum Khan
- grid.10423.340000 0000 9529 9877Laboratory for Molecular Neurosciences (LMN), Department of Psychiatry, Social Psychiatry and Psychotherapy, Medical School Hannover (MHH), Carl-Neuberg-Str. 1, 30625 Hannover, Germany ,grid.444940.9University of Management and Technology—School of Pharmacy, 72-A Raiwind Rd, Dubai Town, Lahore Pakistan
| | - Lukas Thielen
- grid.10423.340000 0000 9529 9877Laboratory for Molecular Neurosciences (LMN), Department of Psychiatry, Social Psychiatry and Psychotherapy, Medical School Hannover (MHH), Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Gwenaëlle Le Pen
- grid.512035.0Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM, Pathophysiology of Psychiatric disorders: Development and Vulnerability, U1266, 102-108 Rue de la Santé, 75014 Paris, France
| | - Marie-Odile Krebs
- grid.512035.0Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM, Pathophysiology of Psychiatric disorders: Development and Vulnerability, U1266, 102-108 Rue de la Santé, 75014 Paris, France ,GHU Paris Psychiatrie et Neurosciences, 1 Rue Cabanis, 75014 Paris, France
| | - Oussama Kebir
- grid.512035.0Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM, Pathophysiology of Psychiatric disorders: Development and Vulnerability, U1266, 102-108 Rue de la Santé, 75014 Paris, France ,GHU Paris Psychiatrie et Neurosciences, 1 Rue Cabanis, 75014 Paris, France
| | - Adrian Groh
- grid.10423.340000 0000 9529 9877Laboratory for Molecular Neurosciences (LMN), Department of Psychiatry, Social Psychiatry and Psychotherapy, Medical School Hannover (MHH), Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Maximilian Deest
- grid.10423.340000 0000 9529 9877Laboratory for Molecular Neurosciences (LMN), Department of Psychiatry, Social Psychiatry and Psychotherapy, Medical School Hannover (MHH), Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Stefan Bleich
- grid.10423.340000 0000 9529 9877Laboratory for Molecular Neurosciences (LMN), Department of Psychiatry, Social Psychiatry and Psychotherapy, Medical School Hannover (MHH), Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Helge Frieling
- grid.10423.340000 0000 9529 9877Laboratory for Molecular Neurosciences (LMN), Department of Psychiatry, Social Psychiatry and Psychotherapy, Medical School Hannover (MHH), Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Kirsten Jahn
- grid.10423.340000 0000 9529 9877Laboratory for Molecular Neurosciences (LMN), Department of Psychiatry, Social Psychiatry and Psychotherapy, Medical School Hannover (MHH), Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
8
|
Hribkova H, Svoboda O, Bartecku E, Zelinkova J, Horinkova J, Lacinova L, Piskacek M, Lipovy B, Provaznik I, Glover JC, Kasparek T, Sun YM. Clozapine Reverses Dysfunction of Glutamatergic Neurons Derived From Clozapine-Responsive Schizophrenia Patients. Front Cell Neurosci 2022; 16:830757. [PMID: 35281293 PMCID: PMC8904748 DOI: 10.3389/fncel.2022.830757] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/28/2022] [Indexed: 11/26/2022] Open
Abstract
The cellular pathology of schizophrenia and the potential of antipsychotics to target underlying neuronal dysfunctions are still largely unknown. We employed glutamatergic neurons derived from induced pluripotent stem cells (iPSC) obtained from schizophrenia patients with known histories of response to clozapine and healthy controls to decipher the mechanisms of action of clozapine, spanning from molecular (transcriptomic profiling) and cellular (electrophysiology) levels to observed clinical effects in living patients. Glutamatergic neurons derived from schizophrenia patients exhibited deficits in intrinsic electrophysiological properties, synaptic function and network activity. Deficits in K+ and Na+ currents, network behavior, and glutamatergic synaptic signaling were restored by clozapine treatment, but only in neurons from clozapine-responsive patients. Moreover, neurons from clozapine-responsive patients exhibited a reciprocal dysregulation of gene expression, particularly related to glutamatergic and downstream signaling, which was reversed by clozapine treatment. Only neurons from clozapine responders showed return to normal function and transcriptomic profile. Our results underscore the importance of K+ and Na+ channels and glutamatergic synaptic signaling in the pathogenesis of schizophrenia and demonstrate that clozapine might act by normalizing perturbances in this signaling pathway. To our knowledge this is the first study to demonstrate that schizophrenia iPSC-derived neurons exhibit a response phenotype correlated with clinical response to an antipsychotic. This opens a new avenue in the search for an effective treatment agent tailored to the needs of individual patients.
Collapse
Affiliation(s)
- Hana Hribkova
- Department of Biology, Masaryk University, Brno, Czechia
| | - Ondrej Svoboda
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Brno, Czechia
| | - Elis Bartecku
- Department of Psychiatry, Faculty of Medicine and University Hospital Brno, Brno, Czechia
| | - Jana Zelinkova
- Department of Biology, Masaryk University, Brno, Czechia
| | - Jana Horinkova
- Department of Psychiatry, Faculty of Medicine and University Hospital Brno, Brno, Czechia
| | - Lubica Lacinova
- Center of Bioscience, Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Martin Piskacek
- Department of Pathological Physiology, Masaryk University, Brno, Czechia
| | - Bretislav Lipovy
- Department of Burns and Plastic Surgery, Faculty of Medicine and University Hospital Brno, Brno, Czechia
| | - Ivo Provaznik
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Brno, Czechia
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Joel C. Glover
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Norwegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital, Oslo, Norway
| | - Tomas Kasparek
- Department of Psychiatry, Faculty of Medicine and University Hospital Brno, Brno, Czechia
- *Correspondence: Tomas Kasparek,
| | - Yuh-Man Sun
- Department of Biology, Masaryk University, Brno, Czechia
| |
Collapse
|
9
|
Aberrant maturation and connectivity of prefrontal cortex in schizophrenia-contribution of NMDA receptor development and hypofunction. Mol Psychiatry 2022; 27:731-743. [PMID: 34163013 PMCID: PMC8695640 DOI: 10.1038/s41380-021-01196-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 02/06/2023]
Abstract
The neurobiology of schizophrenia involves multiple facets of pathophysiology, ranging from its genetic basis over changes in neurochemistry and neurophysiology, to the systemic level of neural circuits. Although the precise mechanisms associated with the neuropathophysiology remain elusive, one essential aspect is the aberrant maturation and connectivity of the prefrontal cortex that leads to complex symptoms in various stages of the disease. Here, we focus on how early developmental dysfunction, especially N-methyl-D-aspartate receptor (NMDAR) development and hypofunction, may lead to the dysfunction of both local circuitry within the prefrontal cortex and its long-range connectivity. More specifically, we will focus on an "all roads lead to Rome" hypothesis, i.e., how NMDAR hypofunction during development acts as a convergence point and leads to local gamma-aminobutyric acid (GABA) deficits and input-output dysconnectivity in the prefrontal cortex, which eventually induce cognitive and social deficits. Many outstanding questions and hypothetical mechanisms are listed for future investigations of this intriguing hypothesis that may lead to a better understanding of the aberrant maturation and connectivity associated with the prefrontal cortex.
Collapse
|
10
|
Sun D, Guo H, Womer FY, Yang J, Tang J, Liu J, Zhu Y, Duan J, Peng Z, Wang H, Tan Q, Zhu Q, Wei Y, Xu K, Zhang Y, Tang Y, Zhang X, Xu F, Wang J, Wang F. Frontal-posterior functional imbalance and aberrant function developmental patterns in schizophrenia. Transl Psychiatry 2021; 11:495. [PMID: 34580274 PMCID: PMC8476507 DOI: 10.1038/s41398-021-01617-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/28/2021] [Accepted: 08/20/2021] [Indexed: 12/01/2022] Open
Abstract
Schizophrenia (SZ) is a neurodevelopmental disorder. There remain significant gaps in understanding the neural trajectory across development in SZ. A major research focus is to clarify the developmental functional changes of SZ and to identify the specific timing, the specific brain regions, and the underlying mechanisms of brain alterations during SZ development. Regional homogeneity (ReHo) characterizing brain function was collected and analyzed on humans with SZ (hSZ) and healthy controls (HC) cross-sectionally, and methylazoxymethanol acetate (MAM) rats, a neurodevelopmental model of SZ, and vehicle rats longitudinally from adolescence to adulthood. Metabolomic and proteomic profiling in adult MAM rats and vehicle rats was examined and bioanalyzed. Compared to HC or adult vehicle rats, similar ReHo alterations were observed in hSZ and adult MAM rats, characterized by increased frontal (medial prefrontal and orbitofrontal cortices) and decreased posterior (visual and associated cortices) ReHo. Longitudinal analysis of MAM rats showed aberrant ReHo patterns as decreased posterior ReHo in adolescence and increased frontal and decreased posterior ReHo in adulthood. Accordingly, it was suggested that the visual cortex was a critical locus and adolescence was a sensitive window in SZ development. In addition, metabolic and proteomic alterations in adult MAM rats suggested that central carbon metabolism disturbance and mitochondrial dysfunction were the potential mechanisms underlying the ReHo alterations. This study proposed frontal-posterior functional imbalance and aberrant function developmental patterns in SZ, suggesting that the adolescent visual cortex was a critical locus and a sensitive window in SZ development. These findings from linking data between hSZ and MAM rats may have a significant translational contribution to the development of effective therapies in SZ.
Collapse
Affiliation(s)
- Dandan Sun
- grid.452816.c0000 0004 1757 9522Department of Cardiovascular Ultrasound, The People’s Hospital of China Medical University & The People’s Hospital of Liaoning Province, Shenyang, China ,grid.412636.4Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
| | - Huiling Guo
- grid.412636.4Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China ,grid.89957.3a0000 0000 9255 8984Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Fay Y. Womer
- grid.4367.60000 0001 2355 7002Department of Psychiatry, Washington University School of Medicine, St Louis, MO USA
| | - Jingyu Yang
- grid.89957.3a0000 0000 9255 8984Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Jingwei Tang
- grid.412636.4Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
| | - Juan Liu
- grid.412636.4Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China ,grid.89957.3a0000 0000 9255 8984Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Yue Zhu
- grid.412636.4Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
| | - Jia Duan
- grid.412636.4Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China ,grid.89957.3a0000 0000 9255 8984Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Zhengwu Peng
- grid.233520.50000 0004 1761 4404Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Huaning Wang
- grid.233520.50000 0004 1761 4404Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Qingrong Tan
- grid.233520.50000 0004 1761 4404Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Qiwen Zhu
- grid.415680.e0000 0000 9549 5392Liaoning Key Laboratory of Cognitive Neuroscience, Shenyang Medical College, Shenyang, China
| | - Yange Wei
- grid.89957.3a0000 0000 9255 8984Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Ke Xu
- grid.412636.4Department of Radiology, The First Hospital of China Medical University, Shenyang, China
| | - Yanbo Zhang
- grid.17089.37Department of Psychiatry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Yanqing Tang
- grid.412636.4Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
| | - Xizhe Zhang
- grid.89957.3a0000 0000 9255 8984School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Fuqiang Xu
- grid.9227.e0000000119573309Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China ,grid.9227.e0000000119573309Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jie Wang
- grid.9227.e0000000119573309Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Fei Wang
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China. .,Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China. .,Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
11
|
Roles of the Functional Interaction between Brain Cholinergic and Dopaminergic Systems in the Pathogenesis and Treatment of Schizophrenia and Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22094299. [PMID: 33919025 PMCID: PMC8122651 DOI: 10.3390/ijms22094299] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/14/2022] Open
Abstract
Most physiologic processes in the brain and related diseases involve more than one neurotransmitter system. Thus, elucidation of the interaction between different neurotransmitter systems could allow for better therapeutic approaches to the treatments of related diseases. Dopaminergic (DAergic) and cholinergic neurotransmitter system regulate various brain functions that include cognition, movement, emotion, etc. This review focuses on the interaction between the brain DAergic and cholinergic systems with respect to the pathogenesis and treatment of schizophrenia and Parkinson’s disease (PD). We first discussed the selection of motor plans at the level of basal ganglia, the major DAergic and cholinergic pathways in the brain, and the receptor subtypes involved in the interaction between the two signaling systems. Next, the roles of each signaling system were discussed in the context of the negative symptoms of schizophrenia, with a focus on the α7 nicotinic cholinergic receptor and the dopamine D1 receptor in the prefrontal cortex. In addition, the roles of the nicotinic and dopamine receptors were discussed in the context of regulation of striatal cholinergic interneurons, which play crucial roles in the degeneration of nigrostriatal DAergic neurons and the development of L-DOPA-induced dyskinesia in PD patients. Finally, we discussed the general mechanisms of nicotine-induced protection of DAergic neurons.
Collapse
|
12
|
Smail MA, Reigle JK, McCullumsmith RE. Using protein turnover to expand the applications of transcriptomics. Sci Rep 2021; 11:4403. [PMID: 33623108 PMCID: PMC7902815 DOI: 10.1038/s41598-021-83886-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 02/08/2021] [Indexed: 01/31/2023] Open
Abstract
RNA expression and protein abundance are often at odds when measured in parallel, raising questions about the functional implications of transcriptomics data. Here, we present the concept of persistence, which attempts to address this challenge by combining protein half-life data with RNA expression into a single metric that approximates protein abundance. The longer a protein's half-life, the more influence it can have on its surroundings. This data offers a valuable opportunity to gain deeper insight into the functional meaning of transcriptome changes. We demonstrate the application of persistence using schizophrenia (SCZ) datasets, where it greatly improved our ability to predict protein abundance from RNA expression. Furthermore, this approach successfully identified persistent genes and pathways known to have impactful changes in SCZ. These results suggest that persistence is a valuable metric for improving the functional insight offered by transcriptomics data, and extended application of this concept could advance numerous research fields.
Collapse
Affiliation(s)
- Marissa A Smail
- Department of Pharmacology and Systems Physiology, University of Cincinnati, 2170 E. Galbraith Rd. Bldg E. Room 216, Cincinnati, OH, 45237-0506, USA.
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA.
| | - James K Reigle
- Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Robert E McCullumsmith
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
- Neurosciences Institute, ProMedica, Toledo, OH, USA
| |
Collapse
|
13
|
Al-Absi AR, Qvist P, Glerup S, Sanchez C, Nyengaard JR. Df(h15q13)/+ Mouse Model Reveals Loss of Astrocytes and Synaptic-Related Changes of the Excitatory and Inhibitory Circuits in the Medial Prefrontal Cortex. Cereb Cortex 2021; 31:1609-1621. [PMID: 33123721 DOI: 10.1093/cercor/bhaa313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 08/19/2020] [Accepted: 09/20/2020] [Indexed: 11/13/2022] Open
Abstract
The 15q13.3 deletion is associated with multiple neurodevelopmental disorders including epilepsy, schizophrenia, and autism. The Df(h15q13)/+ mouse model was recently generated that recapitulates several phenotypic features of the human 15q13.3 deletion syndrome (DS). However, the biological substrates underlying these phenotypes in Df(h15q13)/+ mice have not yet been fully characterized. RNA sequencing followed by real-time quantitative PCR, western blotting, liquid chromatography-mass spectrometry, and stereological analysis were employed to dissect the molecular, structural, and neurochemical phenotypes of the medial prefrontal cortex (mPFC) circuits in Df(h15q13)/+ mouse model. Transcriptomic profiling revealed enrichment for astrocyte-specific genes among differentially expressed genes, translated by a decrease in the number of glial fibrillary acidic protein positive cells in mPFC of Df(h15q13)/+ mice compared with wild-type mice. mPFC in Df(h15q13)/+ mice also showed a deficit of the inhibitory presynaptic marker GAD65, in addition to a reduction in dendritic arborization and spine density of pyramidal neurons from layers II/III. mPFC levels of GABA and glutamate neurotransmitters were not different between genotypes. Our results suggest that the 15q13.3 deletion modulates nonneuronal circuits in mPFC and confers molecular and morphometric alterations in the inhibitory and excitatory neurocircuits, respectively. These alterations potentially contribute to the phenotypes accompanied with the 15q13.3DS.
Collapse
Affiliation(s)
- Abdel-Rahman Al-Absi
- Center for Molecular Morphology, Section for Stereology and Microscopy, Center for Stochastic Geometry and Advanced Bioimaging, Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Per Qvist
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark.,The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, 8210 Aarhus, Denmark.,Centre for Integrative Sequencing, iSEQ, Aarhus University, 8000 Aarhus, Denmark.,Center for Genomics and Personalized Medicine, CGPM, Aarhus University, 8000 Aarhus, Denmark
| | - Simon Glerup
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Connie Sanchez
- Translational Neuropsychiatry Unit, Aarhus University, 8000 Aarhus, Denmark
| | - Jens R Nyengaard
- Center for Molecular Morphology, Section for Stereology and Microscopy, Center for Stochastic Geometry and Advanced Bioimaging, Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
14
|
Ketamine disrupts naturalistic coding of working memory in primate lateral prefrontal cortex networks. Mol Psychiatry 2021; 26:6688-6703. [PMID: 33981008 PMCID: PMC8760073 DOI: 10.1038/s41380-021-01082-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 03/09/2021] [Accepted: 03/26/2021] [Indexed: 01/23/2023]
Abstract
Ketamine is a dissociative anesthetic drug, which has more recently emerged as a rapid-acting antidepressant. When acutely administered at subanesthetic doses, ketamine causes cognitive deficits like those observed in patients with schizophrenia, including impaired working memory. Although these effects have been linked to ketamine's action as an N-methyl-D-aspartate receptor antagonist, it is unclear how synaptic alterations translate into changes in brain microcircuit function that ultimately influence cognition. Here, we administered ketamine to rhesus monkeys during a spatial working memory task set in a naturalistic virtual environment. Ketamine induced transient working memory deficits while sparing perceptual and motor skills. Working memory deficits were accompanied by decreased responses of fast spiking inhibitory interneurons and increased responses of broad spiking excitatory neurons in the lateral prefrontal cortex. This translated into a decrease in neuronal tuning and information encoded by neuronal populations about remembered locations. Our results demonstrate that ketamine differentially affects neuronal types in the neocortex; thus, it perturbs the excitation inhibition balance within prefrontal microcircuits and ultimately leads to selective working memory deficits.
Collapse
|
15
|
Datta D, Enwright JF, Arion D, Paspalas CD, Morozov YM, Lewis DA, Arnsten AFT. Mapping Phosphodiesterase 4D (PDE4D) in Macaque Dorsolateral Prefrontal Cortex: Postsynaptic Compartmentalization in Layer III Pyramidal Cell Circuits. Front Neuroanat 2020; 14:578483. [PMID: 33328902 PMCID: PMC7714912 DOI: 10.3389/fnana.2020.578483] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/06/2020] [Indexed: 12/20/2022] Open
Abstract
cAMP signaling has powerful, negative effects on cognitive functions of the primate dorsolateral prefrontal cortex (dlPFC), opening potassium channels to reduce firing and impair working memory, and increasing tau phosphorylation in aging neurons. This contrasts with cAMP actions in classic circuits, where it enhances plasticity and transmitter release. PDE4 isozymes regulate cAMP actions, and thus have been a focus of research and drug discovery. Previous work has focused on the localization of PDE4A and PDE4B in dlPFC, but PDE4D is also of great interest, as it is the predominant PDE4 isoform in primate association cortex, and PDE4D expression decreases with aging in human dlPFC. Here we used laser-capture microdissection transcriptomics and found that PDE4D message is enriched in pyramidal cells compared to GABAergic PV-interneurons in layer III of the human dlPFC. A parallel study in rhesus macaques using high-spatial resolution immunoelectron microscopy revealed the ultrastructural locations of PDE4D in primate dlPFC with clarity not possible in human post-mortem tissue. PDE4D was especially prominent in dendrites associated with microtubules, mitochondria, and likely smooth endoplasmic reticulum (SER). There was substantial postsynaptic labeling in dendritic spines, associated with the SER spine-apparatus near glutamatergic-like axospinous synapses, but sparse labeling in axon terminals. We also observed dense PDE4D labeling perisynaptically in astroglial leaflets ensheathing glutamatergic connections. These data suggest that PDE4D is strategically positioned to regulate cAMP signaling in dlPFC glutamatergic synapses and circuits, especially in postsynaptic compartments where it is localized to influence cAMP actions on intracellular trafficking, mitochondrial physiology, and internal calcium release.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - John F. Enwright
- Department of Psychiatry, Translational Neuroscience Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Dominique Arion
- Department of Psychiatry, Translational Neuroscience Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Constantinos D. Paspalas
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Yury M. Morozov
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - David A. Lewis
- Department of Psychiatry, Translational Neuroscience Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Amy F. T. Arnsten
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
16
|
Zink CF, Giegerich M, Prettyman GE, Carta KE, van Ginkel M, O'Rourke MP, Singh E, Fuchs EJ, Hendrix CW, Zimmerman E, Breakey J, Marzinke MA, Hummert P, Pillai JJ, Weinberger DR, Bigos KL. Nimodipine improves cortical efficiency during working memory in healthy subjects. Transl Psychiatry 2020; 10:372. [PMID: 33139710 PMCID: PMC7606375 DOI: 10.1038/s41398-020-01066-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 06/11/2020] [Accepted: 06/18/2020] [Indexed: 02/01/2023] Open
Abstract
The L-type calcium channel gene, CACNA1C, is a validated risk gene for schizophrenia and the target of calcium channel blockers. Carriers of the risk-associated genotype (rs1006737 A allele) have increased frontal cortical activity during working memory and higher CACNA1C mRNA expression in the prefrontal cortex. The aim of this study was to determine how the brain-penetrant calcium channel blocker, nimodipine, changes brain activity during working memory and other cognitive and emotional processes. We conducted a double-blind randomized cross-over pharmacoMRI study of a single 60 mg dose of oral nimodipine solution and matching placebo in healthy men, prospectively genotyped for rs1006737. With performance unchanged, nimodipine significantly decreased frontal cortical activity by 39.1% and parietal cortical activity by 42.8% during the N-back task (2-back > 0-back contrast; PFWE < 0.05; n = 28). Higher peripheral nimodipine concentrations were correlated with a greater decrease in activation in the frontal cortex. Carriers of the risk-associated allele, A (n = 14), had a greater decrease in frontal cortical activation during working memory compared to non-risk allele carriers. No differences in brain activation were found between nimodipine and placebo for other tasks. Future studies should be conducted to test if the decreased cortical brain activity after nimodipine is associated with improved working memory performance in patients with schizophrenia, particularly those who carry the risk-associated genotype. Furthermore, changes in cortical activity during working memory may be a useful biomarker in future trials of L-type calcium channel blockers.
Collapse
Affiliation(s)
- Caroline F Zink
- Baltimore Research and Education Foundation, Baltimore, MD, United States
- Lieber Institute for Brain Development, Baltimore, MD, United States
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Mellissa Giegerich
- Lieber Institute for Brain Development, Baltimore, MD, United States
- Veterans Administration, San Diego, CA, United States
| | - Greer E Prettyman
- Lieber Institute for Brain Development, Baltimore, MD, United States
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, United States
| | - Kayla E Carta
- Lieber Institute for Brain Development, Baltimore, MD, United States
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Marcus van Ginkel
- Lieber Institute for Brain Development, Baltimore, MD, United States
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Molly P O'Rourke
- Lieber Institute for Brain Development, Baltimore, MD, United States
- School of Nursing, University of Pennsylvania, Philadelphia, PA, United States
| | - Eesha Singh
- Lieber Institute for Brain Development, Baltimore, MD, United States
- College of Medicine, University of Tennessee, Memphis, TN, United States
| | - Edward J Fuchs
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Craig W Hendrix
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Pharmacology and Molecular Science, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Epidemiology, Johns Hopkins School of Public Health, Baltimore, MD, United States
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Eric Zimmerman
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Jennifer Breakey
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Mark A Marzinke
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Pamela Hummert
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Jay J Pillai
- Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Baltimore, MD, United States
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, United States
- The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Kristin L Bigos
- Lieber Institute for Brain Development, Baltimore, MD, United States.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States.
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD, United States.
- Department of Pharmacology and Molecular Science, Johns Hopkins School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
17
|
Feng G, Jensen FE, Greely HT, Okano H, Treue S, Roberts AC, Fox JG, Caddick S, Poo MM, Newsome WT, Morrison JH. Opportunities and limitations of genetically modified nonhuman primate models for neuroscience research. Proc Natl Acad Sci U S A 2020; 117:24022-24031. [PMID: 32817435 PMCID: PMC7533691 DOI: 10.1073/pnas.2006515117] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The recently developed new genome-editing technologies, such as the CRISPR/Cas system, have opened the door for generating genetically modified nonhuman primate (NHP) models for basic neuroscience and brain disorders research. The complex circuit formation and experience-dependent refinement of the human brain are very difficult to model in vitro, and thus require use of in vivo whole-animal models. For many neurodevelopmental and psychiatric disorders, abnormal circuit formation and refinement might be at the center of their pathophysiology. Importantly, many of the critical circuits and regional cell populations implicated in higher human cognitive function and in many psychiatric disorders are not present in lower mammalian brains, while these analogous areas are replicated in NHP brains. Indeed, neuropsychiatric disorders represent a tremendous health and economic burden globally. The emerging field of genetically modified NHP models has the potential to transform our study of higher brain function and dramatically facilitate the development of effective treatment for human brain disorders. In this paper, we discuss the importance of developing such models, the infrastructure and training needed to maximize the impact of such models, and ethical standards required for using these models.
Collapse
Affiliation(s)
- Guoping Feng
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139;
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142
| | - Frances E Jensen
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104;
| | - Henry T Greely
- Center for Law and the Biosciences, Stanford University, Stanford, CA 94305
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Shinjukuku, 160-8592 Tokyo, Japan
- Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, 351-0106 Saitama, Wakoshi, Japan
| | - Stefan Treue
- Cognitive Neuroscience Laboratory, German Primate Center-Leibniz Institute for Primate Research, 37077 Goettingen, Germany
- Faculty of Biology and Psychology, University of Goettingen, 37073 Goettingen, Germany
| | - Angela C Roberts
- Department of Physiology, Development, and Neuroscience, University of Cambridge, CB2 3DY Cambridge, United Kingdom
| | - James G Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Sarah Caddick
- The Gatsby Charitable Foundation, SW1V 1AP London, United Kingdom
| | - Mu-Ming Poo
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 200031 Shanghai, China
| | - William T Newsome
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA 94305;
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305
| | - John H Morrison
- California National Primate Research Center, University of California, Davis, CA 95616;
- Department of Neurology, School of Medicine, University of California, Davis, CA 95616
| |
Collapse
|
18
|
Werner AL, Tebartz van Elst L, Ebert D, Friedel E, Bubl A, Clement HW, Lukačin R, Bach M, Bubl E. Normalization of increased retinal background noise after ADHD treatment: A neuronal correlate. Schizophr Res 2020; 219:77-83. [PMID: 31053491 DOI: 10.1016/j.schres.2019.04.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/14/2019] [Accepted: 04/17/2019] [Indexed: 02/06/2023]
Abstract
Inattention, distractibility, and problems inhibiting irrelevant information impose a large disease burden in attention deficit hyperactivity disorder (ADHD). Problems with cognitive function are found in many major psychiatric disorders, and our understanding of ADHD might add to our knowledge of other neuropsychiatric disorders. Despite the high impact of ADHD, the pathophysiology and the mechanism of treatment action remains poorly understood. Increasing evidence suggests that elevated neuronal and retinal background noise plays a prominent role in ADHD. However, the effect of treatment (e.g., methylphenidate) on noise remains unclear. For this study, retinal background noise was assessed with the pattern-electroretinogram (PERG) in 20 drug-naïve adults with ADHD before and after treatment with methylphenidate and in 21 control subjects. Background noise was identified using the Fourier magnitude at frequencies adjacent to the stimulus-response frequency of 12.5 Hz. At baseline, we found an elevated retinal background noise in ADHD patients (Mdn = 0.079 μV) compared to controls (Mdn = 0.062 μV; z = -2.79, p = 0.016, r = -0.44). The noise in the ADHD group decreased significantly at follow-up after treatment with methylphenidate (Mdn = 0.069 μV, z = -2.39, p = 0.035, r = -0.39) while there was no change in the control group. PERG-based retinal noise is increased in adult ADHD and normalizes along with clinical symptoms following treatment with methylphenidate. The retinal noise level might be a promising marker of ADHD in clinical and basic research and illustrates the biological match with nonhuman ADHD models.
Collapse
Affiliation(s)
- Anna Lisa Werner
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Germany
| | - Ludger Tebartz van Elst
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Germany
| | - Dieter Ebert
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Germany
| | - Evelyn Friedel
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Germany
| | - Anna Bubl
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Germany; Department of Psychiatry and Psychotherapy, University of Saarland, Germany
| | - Hans-Willi Clement
- Department of Child and Adolescent Psychiatry, Medical Center - University of Freiburg, Germany
| | - Richard Lukačin
- Chromsystems Instruments & Chemicals GmbH, Gräfelfing, Munich, Germany
| | - Michael Bach
- Eye Center, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Emanuel Bubl
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Germany; Department of Psychiatry and Psychotherapy, University of Saarland, Germany.
| |
Collapse
|
19
|
Blockage of NMDA- and GABA(A) Receptors Improves Working Memory Selectivity of Primate Prefrontal Neurons. J Neurosci 2020; 40:1527-1537. [PMID: 31911457 DOI: 10.1523/jneurosci.2009-19.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/03/2019] [Accepted: 12/05/2019] [Indexed: 12/16/2022] Open
Abstract
The ongoing activity of prefrontal neurons after a stimulus has disappeared is considered a neuronal correlate of working memory. It depends on the delicate but poorly understood interplay between excitatory glutamatergic and inhibitory GABAergic receptor effects. We administered the NMDA receptor antagonist MK-801 and the GABA(A) receptor antagonist bicuculline methiodide while recording cellular activity in PFC of male rhesus monkeys performing a delayed decision task requiring working memory. The blockade of GABA(A) receptors strongly improved the selectivity of the neurons' delay activity, causing an increase in signal-to-noise ratio during working memory periods as well as an enhancement of the neurons' coding selectivity. The blockade of NMDA receptors resulted in a slight enhancement of selectivity and encoding capacity of the neurons. Our findings emphasize the delicate and more complex than expected interplay of excitatory and inhibitory transmitter systems in modulating working memory coding in prefrontal circuits.SIGNIFICANCE STATEMENT Ongoing delay activity of prefrontal neurons constitutes a neuronal correlate of working memory. However, how this delay activity is generated by the delicate interplay of synaptic excitation and inhibition is unknown. We probed the effects of excitatory neurotransmitter glutamate and inhibitory neurotransmitter GABA in regulating delay activity in rhesus monkeys performing a delayed decision task requiring working memory. Surprisingly, the blockade of both glutamatergic NMDA and GABA(A) receptors improved neuronal selectivity of delay activity, causing an increase in neuronal signal-to-noise ratio. Moreover, individual neurons were similarly affected by blockade of both receptors. This emphasizes the delicate and more complex than expected interplay of excitatory and inhibitory transmitter systems in modulating working memory coding in prefrontal circuits.
Collapse
|
20
|
Chung YS, Poppe A, Novotny S, Epperson CN, Kober H, Granger DA, Blumberg HP, Ochsner K, Gross JJ, Pearlson G, Stevens MC. A preliminary study of association between adolescent estradiol level and dorsolateral prefrontal cortex activity during emotion regulation. Psychoneuroendocrinology 2019; 109:104398. [PMID: 31394491 PMCID: PMC6842698 DOI: 10.1016/j.psyneuen.2019.104398] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 07/24/2019] [Accepted: 07/24/2019] [Indexed: 12/19/2022]
Abstract
Non-human primate models have been useful in clarifying estradiol's role in cognitive processing. These animal studies indicate estradiol impacts cognitive processes supported by regions within dorsolateral prefrontal cortex (DLPFC). Although human functional neuroimaging studies have begun to find similar relationships between estradiol in women for some forms of 'cold' cognitive control, to date no studies have examined the relationship between estradiol and DLPFC function in the context of active attempts to regulate one's emotions. Here, we asked whether peripheral 17-beta estradiol levels in adolescent girls in different pubertal developmental stages (age = 14.9 years ± 1.74) were related to engagement of DLPFC regions during the use of a cognitive strategy for regulating emotion known as reappraisal using functional Magnetic Resonance Imaging. Findings indicated that higher estradiol levels predicted greater DLPFC activity during the down-regulation of negative emotion using reappraisal. This is the first report of an association between estradiol level and DLPFC activity during cognitive reappraisal of negative emotion. The study suggests a possibility that estradiol might positively contribute to regulatory function of a cortical system important for emotional experiences.
Collapse
Affiliation(s)
- Yu Sun Chung
- Olin Neuropsychiatry Research Center, 200 Retreat Avenue, Whitehall Building- Institute of Living, Hartford, CT, 06106, USA; Department of Psychology and Neuroscience, Duke University.
| | - Andrew Poppe
- Olin Neuropsychiatry Research Center, 200 Retreat Avenue, Whitehall Building- Institute of Living, Hartford, CT, 06106, USA; Department of Psychology and Neuroscience, Duke University
| | - Stephanie Novotny
- Olin Neuropsychiatry Research Center, 200 Retreat Avenue, Whitehall Building- Institute of Living, Hartford, CT, 06106, USA; Department of Psychology and Neuroscience, Duke University
| | - C. Neill Epperson
- Department of Psychiatry, University of Colorado School of Medicine, Aurora, CO, USA
| | - Hedy Kober
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Douglas A. Granger
- Institute for Interdisciplinary Salivary Bioscience Research, University of California at Irvine, Irvine CA; School of Medicine, Bloomberg School of Public Health, and School of Nursing, Johns Hopkins University, Baltimore, MD, USA
| | | | - Kevin Ochsner
- Department of Psychology, Columbia University, New York, NY, USA
| | - James J. Gross
- Department of Psychology, Stanford University, Stanford, CA, USA
| | - Godfrey Pearlson
- Olin Neuropsychiatry Research Center, 200 Retreat Avenue, Whitehall Building- Institute of Living, Hartford, CT, 06106, USA; Department of Psychology and Neuroscience, Duke University.,Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Michael C. Stevens
- Olin Neuropsychiatry Research Center, 200 Retreat Avenue, Whitehall Building- Institute of Living, Hartford, CT, 06106, USA; Department of Psychology and Neuroscience, Duke University.,Department of Psychiatry, Yale University, New Haven, CT, USA
| |
Collapse
|
21
|
Foster TC. Senescent neurophysiology: Ca 2+ signaling from the membrane to the nucleus. Neurobiol Learn Mem 2019; 164:107064. [PMID: 31394200 DOI: 10.1016/j.nlm.2019.107064] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/29/2019] [Accepted: 08/03/2019] [Indexed: 12/16/2022]
Abstract
The current review provides a historical perspective on the evolution of hypothesized mechanisms for senescent neurophysiology, focused on the CA1 region of the hippocampus, and the relationship of senescent neurophysiology to impaired hippocampal-dependent memory. Senescent neurophysiology involves processes linked to calcium (Ca2+) signaling including an increase in the Ca2+-dependent afterhyperpolarization (AHP), decreasing pyramidal cell excitability, hyporesponsiveness of N-methyl-D-aspartate (NMDA) receptor function, and a shift in Ca2+-dependent synaptic plasticity. Dysregulation of intracellular Ca2+ and downstream signaling of kinase and phosphatase activity lies at the core of senescent neurophysiology. Ca2+-dysregulation involves a decrease in Ca2+ influx through NMDA receptors and an increase release of Ca2+ from internal Ca2+ stores. Recent work has identified changes in redox signaling, arising in middle-age, as an initiating factor for senescent neurophysiology. The shift in redox state links processes of aging, oxidative stress and inflammation, with functional changes in mechanisms required for episodic memory. The link between age-related changes in Ca2+ signaling, epigenetics and gene expression is an exciting area of research. Pharmacological and behavioral intervention, initiated in middle-age, can promote memory function by initiating transcription of neuroprotective genes and rejuvenating neurophysiology. However, with more advanced age, or under conditions of neurodegenerative disease, epigenetic changes may weaken the link between environmental influences and transcription, decreasing resilience of memory function.
Collapse
Affiliation(s)
- Thomas C Foster
- Department of Neuroscience and Genetics and Genomics Program, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
22
|
Datta D, Yang ST, Galvin VC, Solder J, Luo F, Morozov YM, Arellano J, Duque A, Rakic P, Arnsten AFT, Wang M. Noradrenergic α1-Adrenoceptor Actions in the Primate Dorsolateral Prefrontal Cortex. J Neurosci 2019; 39:2722-2734. [PMID: 30755491 PMCID: PMC6445993 DOI: 10.1523/jneurosci.2472-18.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 01/23/2019] [Accepted: 01/28/2019] [Indexed: 01/14/2023] Open
Abstract
Noradrenergic (NE) α1-adrenoceptors (α1-ARs) contribute to arousal mechanisms and play an important role in therapeutic medications such as those for the treatment of posttraumatic stress disorder (PTSD). However, little is known about how α1-AR stimulation influences neuronal firing in the dorsolateral prefrontal cortex (dlPFC), a newly evolved region that is dysfunctional in PTSD and other mental illnesses. The current study examined the effects of α1-AR manipulation on neuronal firing in dlPFC of rhesus monkeys performing a visuospatial working memory task, focusing on the "delay cells" that maintain spatially tuned information across the delay period. Iontophoresis of the α1-AR antagonist HEAT (2-{[β-(4-hydroxyphenyl)ethyl]aminomethyl}-1-tetralone) had mixed effects, reducing firing in a majority of neurons but having nonsignificant excitatory effects or no effect in remaining delay cells. These data suggest that endogenous NE has excitatory effects in some delay cells under basal conditions. In contrast, the α1-AR agonists phenylephrine and cirazoline suppressed delay cell firing and this was blocked by coadministration of HEAT. These results indicate an inverted-U dose response for α1-AR actions, with mixed excitatory actions under basal conditions and suppressed firing with high levels of α1-AR stimulation such as with stress exposure. Immunoelectron microscopy revealed α1-AR expression presynaptically in axons and axon terminals and postsynaptically in spines, dendrites, and astrocytes. It is possible that α1-AR excitatory effects arise from presynaptic excitation of glutamate release, whereas postsynaptic actions suppress firing through calcium-protein kinase C opening of potassium channels on spines. The latter may predominate under stressful conditions, leading to loss of dlPFC regulation during uncontrollable stress.SIGNIFICANCE STATEMENT Noradrenergic stimulation of α1-adrenoceptors (α1-ARs) is implicated in posttraumatic stress disorder (PTSD) and other mental disorders that involve dysfunction of the prefrontal cortex, a brain region that provides top-down control. However, the location and contribution of α1-ARs to prefrontal cortical physiology in primates has received little attention. This study found that α1-ARs are located near prefrontal synapses and that α1-AR stimulation has mixed effects under basal conditions. However, high levels of α1-AR stimulation, as occur with stress, suppress neuronal firing. These findings help to explain why we lose top-down control under conditions of uncontrollable stress when there are high levels of noradrenergic release in brain and why blocking α1-AR, such as with prazosin, may be helpful in the treatment of PTSD.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520, and
| | - Sheng-Tao Yang
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520, and
| | - Veronica C Galvin
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520, and
| | - John Solder
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520, and
| | - Fei Luo
- Center for Neuropsychiatric Diseases, Institute of Life Science, Nanchang University, Nanchang 330031, China
| | - Yury M Morozov
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520, and
| | - Jon Arellano
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520, and
| | - Alvaro Duque
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520, and
| | - Pasko Rakic
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520, and
| | - Amy F T Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520, and
| | - Min Wang
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520, and
| |
Collapse
|
23
|
Petanjek Z, Sedmak D, Džaja D, Hladnik A, Rašin MR, Jovanov-Milosevic N. The Protracted Maturation of Associative Layer IIIC Pyramidal Neurons in the Human Prefrontal Cortex During Childhood: A Major Role in Cognitive Development and Selective Alteration in Autism. Front Psychiatry 2019; 10:122. [PMID: 30923504 PMCID: PMC6426783 DOI: 10.3389/fpsyt.2019.00122] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 02/18/2019] [Indexed: 12/12/2022] Open
Abstract
The human specific cognitive shift starts around the age of 2 years with the onset of self-awareness, and continues with extraordinary increase in cognitive capacities during early childhood. Diffuse changes in functional connectivity in children aged 2-6 years indicate an increase in the capacity of cortical network. Interestingly, structural network complexity does not increase during this time and, thus, it is likely to be induced by selective maturation of a specific neuronal subclass. Here, we provide an overview of a subclass of cortico-cortical neurons, the associative layer IIIC pyramids of the human prefrontal cortex. Their local axonal collaterals are in control of the prefrontal cortico-cortical output, while their long projections modulate inter-areal processing. In this way, layer IIIC pyramids are the major integrative element of cortical processing, and changes in their connectivity patterns will affect global cortical functioning. Layer IIIC neurons have a unique pattern of dendritic maturation. In contrast to other classes of principal neurons, they undergo an additional phase of extensive dendritic growth during early childhood, and show characteristic molecular changes. Taken together, circuits associated with layer IIIC neurons have the most protracted period of developmental plasticity. This unique feature is advanced but also provides a window of opportunity for pathological events to disrupt normal formation of cognitive circuits involving layer IIIC neurons. In this manuscript, we discuss how disrupted dendritic and axonal maturation of layer IIIC neurons may lead into global cortical disconnectivity, affecting development of complex communication and social abilities. We also propose a model that developmentally dictated incorporation of layer IIIC neurons into maturing cortico-cortical circuits between 2 to 6 years will reveal a previous (perinatal) lesion affecting other classes of principal neurons. This "disclosure" of pre-existing functionally silent lesions of other neuronal classes induced by development of layer IIIC associative neurons, or their direct alteration, could be found in different forms of autism spectrum disorders. Understanding the gene-environment interaction in shaping cognitive microcircuitries may be fundamental for developing rehabilitation and prevention strategies in autism spectrum and other cognitive disorders.
Collapse
Affiliation(s)
- Zdravko Petanjek
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Dora Sedmak
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Domagoj Džaja
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ana Hladnik
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Mladen Roko Rašin
- Department of Neuroscience and Cell Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Nataša Jovanov-Milosevic
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Medical Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
24
|
Wang M, Datta D, Enwright J, Galvin V, Yang ST, Paspalas C, Kozak R, Gray DL, Lewis DA, Arnsten AFT. A novel dopamine D1 receptor agonist excites delay-dependent working memory-related neuronal firing in primate dorsolateral prefrontal cortex. Neuropharmacology 2019; 150:46-58. [PMID: 30858103 DOI: 10.1016/j.neuropharm.2019.03.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 02/28/2019] [Accepted: 03/01/2019] [Indexed: 01/10/2023]
Abstract
Decades of research have emphasized the importance of dopamine (DA) D1 receptor (D1R) mechanisms to dorsolateral prefrontal cortex (dlPFC) working memory function, and the hope that D1R agonists could be used to treat cognitive disorders. However, existing D1R agonists all have had high affinity for D1R, and engage β-arrestin signaling, and these agonists have suppressed task-related neuronal firing. The current study provides the first physiological characterization of a novel D1R agonist, PF-3628, with low affinity for D1R -more similar to endogenous DA actions- as well as little engagement of β-arrestin signaling. PF-3628 was applied by iontophoresis directly onto dlPFC neurons in aged rhesus monkeys performing a delay-dependent working memory task. Aged monkeys have naturally-occurring loss of DA, and naturally-occurring reductions in dlPFC neuronal firing and working memory performance. We found the first evidence of excitatory actions of a D1R agonist on dlPFC task-related firing, and this PF-3628 beneficial response was blocked by co-application of a D1R antagonist. These D1R actions likely occur on pyramidal cells, based on previous immunoelectron microscopic studies showing expression of D1R on layer III spines, and current microarray experiments showing that D1R are four times more prevalent in pyramidal cells than in parvalbumin-containing interneurons laser-captured from layer III of the human dlPFC. These results encourage the translation of D1R mechanisms from monkey to human, with the hope PF-3628 and related, novel D1R agonists will be more appropriate for enhancing dlPFC cognitive functions in patients with mental disorders.
Collapse
Affiliation(s)
- Min Wang
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA.
| | - Dibyadeep Datta
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - John Enwright
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Veronica Galvin
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Sheng-Tao Yang
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Constantinos Paspalas
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Rouba Kozak
- Pfizer Inc, Internal Medicine Unit, Pfizer Inc., 1 Portland St., Cambridge, MA, 02139, USA
| | - David L Gray
- Pfizer Inc, Internal Medicine Unit, Pfizer Inc., 1 Portland St., Cambridge, MA, 02139, USA
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Amy F T Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|