1
|
Alabdali AN, Ben Bacha A, Alonazi M, Abuaish S, Almotairi A, Al-Ayadhi L, El-Ansary AK. Impact of GABA and nutritional supplements on neurochemical biomarkers in autism: a PPA rodent model study. Front Mol Neurosci 2025; 18:1553438. [PMID: 40171233 PMCID: PMC11959029 DOI: 10.3389/fnmol.2025.1553438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/03/2025] [Indexed: 04/03/2025] Open
Abstract
Background/objectives Autism spectrum disorder (ASD) is associated with excitatory-inhibitory imbalance and oxidative stress. GABA, an inhibitory neurotransmitter, and related nutritional therapies are promising in restoring these imbalances. GABAergic deficits and glutamate excitotoxicity are two essential signaling pathways that could be addressed to treat autism, thus medications targeting these pathways are critical for treating behavioral symptoms. In a rat model of autism produced by propionic acid (PPA), this study assessed the effects of GABA supplementation and combined nutritional therapy (probiotics, vitamin D3) and β-lactam as an activator of glutamate transporter. Methods Sixty rats were randomly assigned into six groups: Group I (Control), Group II (PPA-treated), Group III (Control-GABA), Group IV (Control-Combination), Group V (PPA-GABA), and Group VI (PPA-Combination). Social behavior was evaluated using the three-chamber test. Selected biochemical variables related to oxidative stress (GST, Catalase, Lipid peroxides, GSH and Vitamin C), GABA and glutamate signaling (EAAT2, KCC2, NKCC1, GABA, VD3, Glutamate and GABRA5) were measured in the brain homogenates of the six groups. The hippocampus was examined histopathologically to assess cellular integrity. Results The obtained data revealed that PPA treatment caused significant oxidative stress and neurotransmitter imbalances, characterized by reduced GABA and elevated glutamate levels. GABA supplementation alone produced moderate benefits in biochemical and behavioral markers, but combined therapy considerably restored GABA levels, reduced oxidative stress, and enhanced social interaction behaviors. Histopathology revealed that combination therapy mitigated neurodegenerative changes induced by PPA, preserving hippocampal cellular structure. Conclusion This study demonstrated that combined therapy (GABA, probiotics, vitamin D3, and β-lactam) were more effective than GABA alone in enhancing neurochemical balance and lowering oxidative stress in a PPA-induced mouse model of autism, indicating promise for treating symptoms.
Collapse
Affiliation(s)
- Altaf N. Alabdali
- Biochemistry Department, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Abir Ben Bacha
- Biochemistry Department, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Mona Alonazi
- Biochemistry Department, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Sameera Abuaish
- Department of Basic Sciences, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Ahmad Almotairi
- Department of Pathology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Laila Al-Ayadhi
- Autism Research and Treatment Center, Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Afaf K. El-Ansary
- Autism Center, Lotus Holistic Medical Center, Abu Dhabi, United Arab Emirates
| |
Collapse
|
2
|
del Cerro-León A, Fernando Antón-Toro L, Shpakivska-Bilan D, Uceta M, Santos-Mayo A, Cuesta P, Bruña R, García-Moreno LM, Maestú F. Adolescent alcohol consumption predicted by differences in electrophysiological functional connectivity and neuroanatomy. Proc Natl Acad Sci U S A 2024; 121:e2320805121. [PMID: 39378092 PMCID: PMC11494299 DOI: 10.1073/pnas.2320805121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 07/22/2024] [Indexed: 10/10/2024] Open
Abstract
Alcohol consumption during adolescence has been associated with neuroanatomical abnormalities and the appearance of future disorders. However, the latest advances in this field point to the existence of risk profiles which may lead to some individuals into an early consumption. To date, some studies have established predictive models of consumption based on sociodemographic, behavioral, and anatomical-functional variables using MRI. However, the neuroimaging variables employed are usually restricted to local and hemodynamic phenomena. Given the potential of connectome approaches, and the high temporal dynamics of electrophysiology, we decided to explore the relationship between future alcohol consumption and electrophysiological connectivity measured by MEG in a cohort of 83 individuals aged 14 to 16. As a result, we found a positive correlation between alcohol consumption and the functional connectivity in frontal, parietal, and frontoparietal connections. Once this relationship was described, multivariate linear regression analyses were used to evaluate the predictive capacity of functional connectivity in conjunction with other neuroanatomical and behavioral variables described in the literature. Finally, the multivariate linear regression analysis determined the importance of anatomical and functional variables in the prediction of alcohol consumption but failed to find associations with impulsivity, sensation seeking, and executive function scales. In conclusion, the predictive traits obtained in these models were closely associated with changes occurring during adolescence, suggesting the existence of different paths in neurodevelopment that have the potential to influence adolescents' relationship with alcohol consumption.
Collapse
Affiliation(s)
- Alberto del Cerro-León
- Center of Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, Madrid28040, Spain
- Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Faculty of Psychology, Universidad Complutense de Madrid, Madrid28040, Spain
| | - Luis Fernando Antón-Toro
- Center of Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, Madrid28040, Spain
- Department of Psychology, University Camilo José Cela, Madrid28692, Spain
| | - Danylyna Shpakivska-Bilan
- Center of Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, Madrid28040, Spain
- Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Faculty of Psychology, Universidad Complutense de Madrid, Madrid28040, Spain
| | - Marcos Uceta
- Center of Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, Madrid28040, Spain
- Department of Cellular Biology, Faculty of Biology, Complutense University of Madrid, Madrid28040, Spain
| | - Alejandro Santos-Mayo
- Center of Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, Madrid28040, Spain
- Department of Psychology, University of Florida, Gainesville, FL32612
| | - Pablo Cuesta
- Center of Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, Madrid28040, Spain
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), Madrid28040, Spain
- Department of Radiology, Faculty of Medicine, Complutense University of Madrid, Madrid28040, Spain
| | - Ricardo Bruña
- Center of Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, Madrid28040, Spain
- Department of Radiology, Faculty of Medicine, Complutense University of Madrid, Madrid28040, Spain
| | - Luis M. García-Moreno
- Department of Psychobiology and Methodology in Behavioral Science, Faculty of Education, Complutense University of Madrid, Madrid28040, Spain
| | - Fernando Maestú
- Center of Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, Madrid28040, Spain
- Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Faculty of Psychology, Universidad Complutense de Madrid, Madrid28040, Spain
- Department of Psychology, University of Florida, Gainesville, FL32612
| |
Collapse
|
3
|
Yao Y, Li Q. The Role of Parvalbumin Interneurons in Autism Spectrum Disorder. J Neurosci Res 2024; 102:e25391. [PMID: 39400385 DOI: 10.1002/jnr.25391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 05/29/2024] [Accepted: 09/29/2024] [Indexed: 10/15/2024]
Abstract
As an important subtype of GABAergic interneurons, parvalbumin (PV) interneurons play a critical role in regulating cortical circuits and neural networks. Abnormalities in the development or function of PV interneurons have been linked to autism spectrum disorder (ASD), a neurodevelopmental disorder characterized by social and language deficits. In this review, we focus on the abnormalities of PV interneurons in ASD, including quantity and function and discuss the underlying mechanisms of impairments in PV interneurons in the pathology of ASD. Finally, we propose potential therapeutic approaches targeting PV interneurons, such as transplanting MGE progenitor cells and utilizing optogenetic stimulation in the treatment of ASD.
Collapse
Affiliation(s)
- Yiwei Yao
- Department of Central Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Qian Li
- Department of Central Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
4
|
Blokland G, Maleki N, Jovicich J, Mesholam-Gately R, DeLisi L, Turner J, Shenton M, Voineskos A, Kahn R, Roffman J, Holt D, Ehrlich S, Kikinis Z, Dazzan P, Murray R, Lee J, Sim K, Lam M, de Zwarte S, Walton E, Kelly S, Picchioni M, Bramon E, Makris N, David A, Mondelli V, Reinders A, Oykhman E, Morris D, Gill M, Corvin A, Cahn W, Ho N, Liu J, Gollub R, Manoach D, Calhoun V, Sponheim S, Buka S, Cherkerzian S, Thermenos H, Dickie E, Ciufolini S, Reis Marques T, Crossley N, Purcell S, Smoller J, van Haren N, Toulopoulou T, Donohoe G, Goldstein J, Keshavan M, Petryshen T, del Re E. MIR137 polygenic risk for schizophrenia and ephrin-regulated pathway: Role in lateral ventricles and corpus callosum volume. Int J Clin Health Psychol 2024; 24:100458. [PMID: 38623146 PMCID: PMC11017057 DOI: 10.1016/j.ijchp.2024.100458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/02/2024] [Indexed: 04/17/2024] Open
Abstract
Background/Objective. Enlarged lateral ventricle (LV) volume and decreased volume in the corpus callosum (CC) are hallmarks of schizophrenia (SZ). We previously showed an inverse correlation between LV and CC volumes in SZ, with global functioning decreasing with increased LV volume. This study investigates the relationship between LV volume, CC abnormalities, and the microRNA MIR137 and its regulated genes in SZ, because of MIR137's essential role in neurodevelopment. Methods. Participants were 1224 SZ probands and 1466 unaffected controls from the GENUS Consortium. Brain MRI scans, genotype, and clinical data were harmonized across cohorts and employed in the analyses. Results. Increased LV volumes and decreased CC central, mid-anterior, and mid-posterior volumes were observed in SZ probands. The MIR137-regulated ephrin pathway was significantly associated with CC:LV ratio, explaining a significant proportion (3.42 %) of CC:LV variance, and more than for LV and CC separately. Other pathways explained variance in either CC or LV, but not both. CC:LV ratio was also positively correlated with Global Assessment of Functioning, supporting previous subsample findings. SNP-based heritability estimates were higher for CC central:LV ratio (0.79) compared to CC or LV separately. Discussion. Our results indicate that the CC:LV ratio is highly heritable, influenced in part by variation in the MIR137-regulated ephrin pathway. Findings suggest that the CC:LV ratio may be a risk indicator in SZ that correlates with global functioning.
Collapse
Affiliation(s)
- G.A.M. Blokland
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Netherlands
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - N. Maleki
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- MGH/HST Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, United States
| | - J. Jovicich
- Center for Mind/Brain Sciences (CIMeC), University of Trento, Trento, Italy
| | - R.I. Mesholam-Gately
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- Massachusetts Mental Health Center Public Psychiatry Division, Beth Israel Deaconess Medical Center, Boston, MA, United States
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - L.E. DeLisi
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- Department of Psychiatry, Cambridge Health Alliance, Cambridge, MA, United States
| | - J.A. Turner
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, United States
| | - M.E. Shenton
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, United States
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Department of Psychiatry, Veterans Affairs Boston Healthcare System, Brockton, MA, United States
| | - A.N. Voineskos
- Kimel Family Translational Imaging Genetics Laboratory, Department of Psychiatry, Research Imaging Centre, Campbell Family Mental Health Institute, Centre for Addiction and Mental Health, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry and Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - R.S. Kahn
- Brain Centre Rudolf Magnus, Department of Psychiatry, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - J.L. Roffman
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- MGH/HST Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, United States
| | - D.J. Holt
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- MGH/HST Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, United States
| | - S. Ehrlich
- Division of Psychological & Social Medicine and Developmental Neurosciences, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Z. Kikinis
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, United States
| | - P. Dazzan
- Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
- National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - R.M. Murray
- Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
- National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - J. Lee
- Institute of Mental Health, Woodbridge Hospital, Singapore
| | - K. Sim
- Institute of Mental Health, Woodbridge Hospital, Singapore
| | - M. Lam
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States
- Institute of Mental Health, Woodbridge Hospital, Singapore
- Analytical & Translational Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States
- Division of Psychiatry Research, The Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY, USA
| | - S.M.C. de Zwarte
- Brain Centre Rudolf Magnus, Department of Psychiatry, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - E. Walton
- Department of Psychology, University of Bath, Bath, United Kingdom
| | - S. Kelly
- Neuropsychiatric Genetics Research Group, Department of Psychiatry, Institute of Molecular Medicine, Trinity College Dublin, Dublin, Ireland
- Laboratory of NeuroImaging, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - M.M. Picchioni
- Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
- National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - E. Bramon
- Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
- National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, United Kingdom
- Mental Health Neuroscience Research Department, UCL Division of Psychiatry, University College London, United Kingdom
| | - N. Makris
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- MGH/HST Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, United States
- Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
- Department of Neurology, Harvard Medical School, Boston, MA, United States
| | - A.S. David
- Division of Psychiatry, University College London, London, United Kingdom
| | - V. Mondelli
- Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
- National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - A.A.T.S. Reinders
- Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
- National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - E. Oykhman
- Massachusetts Mental Health Center Public Psychiatry Division, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - D.W. Morris
- Cognitive Genetics and Cognitive Therapy Group, Neuroimaging and Cognitive Genomics (NICOG) Centre and NCBES Galway Neuroscience Centre, School of Psychology and Discipline of Biochemistry, National University of Ireland, Galway, Ireland
| | - M. Gill
- Neuropsychiatric Genetics Research Group, Department of Psychiatry, Institute of Molecular Medicine, Trinity College Dublin, Dublin, Ireland
| | - A.P. Corvin
- Neuropsychiatric Genetics Research Group, Department of Psychiatry, Institute of Molecular Medicine, Trinity College Dublin, Dublin, Ireland
| | - W. Cahn
- Brain Centre Rudolf Magnus, Department of Psychiatry, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - N. Ho
- Institute of Mental Health, Woodbridge Hospital, Singapore
| | - J. Liu
- Genome Institute, Singapore
| | - R.L. Gollub
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- MGH/HST Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, United States
| | - D.S. Manoach
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- MGH/HST Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, United States
| | - V.D. Calhoun
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State, Georgia Tech, Emory, Atlanta, GA, United States
| | - S.R. Sponheim
- Department of Psychiatry, University of Minnesota, Minneapolis, MN, United States
| | - S.L. Buka
- Department of Epidemiology, Brown University, Providence, RI, United States
| | - S. Cherkerzian
- Department of Medicine, Division of Women's Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - H.W. Thermenos
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- Massachusetts Mental Health Center Public Psychiatry Division, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - E.W. Dickie
- Kimel Family Translational Imaging Genetics Laboratory, Department of Psychiatry, Research Imaging Centre, Campbell Family Mental Health Institute, Centre for Addiction and Mental Health, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - S. Ciufolini
- Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
- National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - T. Reis Marques
- Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
- National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - N.A. Crossley
- Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
- National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - S.M. Purcell
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States
- Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, United States
- Division of Psychiatric Genomics, Departments of Psychiatry and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - J.W. Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - N.E.M. van Haren
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus Medical Centre, Rotterdam, The Netherlands
- Department of Psychiatry, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - T. Toulopoulou
- Department of Psychology & National Magnetic Resonance Research Center (UMRAM), Aysel Sabuncu Brain Research Centre (ASBAM), Bilkent University, Ankara, Turkey
- Department of Psychiatry, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - G. Donohoe
- Neuropsychiatric Genetics Research Group, Department of Psychiatry, Institute of Molecular Medicine, Trinity College Dublin, Dublin, Ireland
- Cognitive Genetics and Cognitive Therapy Group, Neuroimaging and Cognitive Genomics (NICOG) Centre and NCBES Galway Neuroscience Centre, School of Psychology and Discipline of Biochemistry, National University of Ireland, Galway, Ireland
| | - J.M. Goldstein
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- Department of Medicine, Division of Women's Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, United States
| | - M.S. Keshavan
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- Massachusetts Mental Health Center Public Psychiatry Division, Beth Israel Deaconess Medical Center, Boston, MA, United States
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - T.L. Petryshen
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - E.C. del Re
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- Massachusetts Mental Health Center Public Psychiatry Division, Beth Israel Deaconess Medical Center, Boston, MA, United States
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Department of Psychiatry, Veterans Affairs Boston Healthcare System, Brockton, MA, United States
| |
Collapse
|
5
|
Kanigowski D, Urban-Ciecko J. Conditioning and pseudoconditioning differently change intrinsic excitability of inhibitory interneurons in the neocortex. Cereb Cortex 2024; 34:bhae109. [PMID: 38572735 PMCID: PMC10993172 DOI: 10.1093/cercor/bhae109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 04/05/2024] Open
Abstract
Many studies indicate a broad role of various classes of GABAergic interneurons in the processes related to learning. However, little is known about how the learning process affects intrinsic excitability of specific classes of interneurons in the neocortex. To determine this, we employed a simple model of conditional learning in mice where vibrissae stimulation was used as a conditioned stimulus and a tail shock as an unconditioned one. In vitro whole-cell patch-clamp recordings showed an increase in intrinsic excitability of low-threshold spiking somatostatin-expressing interneurons (SST-INs) in layer 4 (L4) of the somatosensory (barrel) cortex after the conditioning paradigm. In contrast, pseudoconditioning reduced intrinsic excitability of SST-LTS, parvalbumin-expressing interneurons (PV-INs), and vasoactive intestinal polypeptide-expressing interneurons (VIP-INs) with accommodating pattern in L4 of the barrel cortex. In general, increased intrinsic excitability was accompanied by narrowing of action potentials (APs), whereas decreased intrinsic excitability coincided with AP broadening. Altogether, these results show that both conditioning and pseudoconditioning lead to plastic changes in intrinsic excitability of GABAergic interneurons in a cell-specific manner. In this way, changes in intrinsic excitability can be perceived as a common mechanism of learning-induced plasticity in the GABAergic system.
Collapse
Affiliation(s)
- Dominik Kanigowski
- Laboratory of Electrophysiology, Nencki Institute of Experimental Biology PAS, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Joanna Urban-Ciecko
- Laboratory of Electrophysiology, Nencki Institute of Experimental Biology PAS, 3 Pasteur Street, 02-093 Warsaw, Poland
| |
Collapse
|
6
|
De Clerck M, Manguin M, Henkous N, d’Almeida MN, Beracochea D, Mons N. Chronic alcohol-induced long-lasting working memory deficits are associated with altered histone H3K9 dimethylation in the prefrontal cortex. Front Behav Neurosci 2024; 18:1354390. [PMID: 38495426 PMCID: PMC10941761 DOI: 10.3389/fnbeh.2024.1354390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/19/2024] [Indexed: 03/19/2024] Open
Abstract
Introduction Epigenetic modifications have emerged as key contributors to the enduring behavioral, molecular and epigenetic neuroadaptations during withdrawal from chronic alcohol exposure. The present study investigated the long-term consequences of chronic alcohol exposure on spatial working memory (WM) and associated changes of transcriptionally repressive histone H3 lysine 9 dimethylation (H3K9me2) in the prefrontal cortex (PFC). Methods Male C57BL/6 mice were allowed free access to either 12% (v/v) ethanol for 5 months followed by a 3-week abstinence period or water. Spatial WM was assessed through the spontaneous alternation T-maze test. Alcoholic and water mice received daily injections of GABAB agonist baclofen or saline during alcohol fading and early withdrawal. Global levels of histone modifications were determined by immunohistochemistry. Results Withdrawal mice displayed WM impairments along with reduced prefrontal H3K9me2 levels, compared to water-drinking mice. The withdrawal-induced decrease of H3K9me2 occurred concomitantly with increased level of permissive H3K9 acetylation (H3K9ac) in the PFC. Baclofen treatment rescued withdrawal-related WM deficits and fully restored prefrontal H3K9me2 and H3K9ac. Alcohol withdrawal induced brain region-specific changes of H3K9me2 and H3K9ac after testing, with significant decreases of both histone marks in the dorsal hippocampus and no changes in the amygdala and dorsal striatum. Furthermore, the magnitude of H3K9me2 in the PFC, but not the hippocampus, significantly and positively correlated with individual WM performances. No correlation was observed between H3K9ac and behavioral performance. Results also indicate that pre-testing intraperitoneal injection of UNC0642, a selective inhibitor of histone methyltransferase G9a responsible for H3K9me2, led to WM impairments in water-drinking and withdrawal-baclofen mice. Collectively, our results demonstrate that alcohol withdrawal induced brain-region specific alterations of H3K9me2 and H3K9ac, an effect that persisted for at least three weeks after cessation of chronic alcohol intake. Conclusion The findings suggest a role for long-lasting decreased H3K9me2 specifically in the PFC in the persistent WM impairments related to alcohol withdrawal.
Collapse
|
7
|
Johnston JN, Kadriu B, Kraus C, Henter ID, Zarate CA. Ketamine in neuropsychiatric disorders: an update. Neuropsychopharmacology 2024; 49:23-40. [PMID: 37340091 PMCID: PMC10700638 DOI: 10.1038/s41386-023-01632-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/08/2023] [Accepted: 06/01/2023] [Indexed: 06/22/2023]
Abstract
The discovery of ketamine as a rapid-acting antidepressant led to a new era in the development of neuropsychiatric therapeutics, one characterized by an antidepressant response that occurred within hours or days rather than weeks or months. Considerable clinical research supports the use of-or further research with-subanesthetic-dose ketamine and its (S)-enantiomer esketamine in multiple neuropsychiatric disorders including depression, bipolar disorder, anxiety spectrum disorders, substance use disorders, and eating disorders, as well as for the management of chronic pain. In addition, ketamine often effectively targets symptom domains associated with multiple disorders, such as anxiety, anhedonia, and suicidal ideation. This manuscript: 1) reviews the literature on the pharmacology and hypothesized mechanisms of subanesthetic-dose ketamine in clinical research; 2) describes similarities and differences in the mechanism of action and antidepressant efficacy between racemic ketamine, its (S) and (R) enantiomers, and its hydroxynorketamine (HNK) metabolite; 3) discusses the day-to-day use of ketamine in the clinical setting; 4) provides an overview of ketamine use in other psychiatric disorders and depression-related comorbidities (e.g., suicidal ideation); and 5) provides insights into the mechanisms of ketamine and therapeutic response gleaned from the study of other novel therapeutics and neuroimaging modalities.
Collapse
Affiliation(s)
- Jenessa N Johnston
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Bashkim Kadriu
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
- Translational and Experimental Medicine, Neuroscience at Jazz Pharmaceuticals, San Diego, CA, USA
| | - Christoph Kraus
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Ioline D Henter
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
8
|
Santos-Silva T, dos Santos Fabris D, de Oliveira CL, Guimarães FS, Gomes FV. Prefrontal and Hippocampal Parvalbumin Interneurons in Animal Models for Schizophrenia: A Systematic Review and Meta-analysis. Schizophr Bull 2024; 50:210-223. [PMID: 37584417 PMCID: PMC10754178 DOI: 10.1093/schbul/sbad123] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
BACKGROUND Consistent with postmortem findings in patients, most animal models for schizophrenia (SCZ) present abnormal levels of parvalbumin (PV), a marker of fast-spiking GABAergic interneurons, in the prefrontal cortex (PFC) and hippocampus (HIP). However, there are discrepancies in the literature. PV reductions lead to a functional loss of PV interneurons, which is proposed to underly SCZ symptoms. Given its complex etiology, different categories of animal models have been developed to study SCZ, which may distinctly impact PV levels in rodent brain areas. STUDY DESIGN We performed a quantitative meta-analysis on PV-positive cell number/density and expression levels in the PFC and HIP of animal models for SCZ based on pharmacological, neurodevelopmental, and genetic manipulations. RESULTS Our results confirmed that PV levels are significantly reduced in the PFC and HIP regardless of the animal model. By categorizing into subgroups, we found that all pharmacological models based on NMDA receptor antagonism decreased PV-positive cell number/density or PV expression levels in both brain areas examined. In neurodevelopmental models, abnormal PV levels were confirmed in both brain areas in maternal immune activation models and HIP of the methylazoxymethanol acetate model. In genetic models, negative effects were found in neuregulin 1 and ERBB4 mutant mice in both brain regions and the PFC of dysbindin mutant mice. Regarding sex differences, male rodents exhibited PV reductions in both brain regions only in pharmacological models, while few studies have been conducted in females. CONCLUSION Overall, our findings support deficits in prefrontal and hippocampal PV interneurons in animal models for SCZ.
Collapse
Affiliation(s)
- Thamyris Santos-Silva
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Débora dos Santos Fabris
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Cilene Lino de Oliveira
- Department of Physiological Sciences, Center of Biological Sciences, University of Santa Catarina, Florianópolis,Brazil
| | - Francisco S Guimarães
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Felipe V Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
9
|
Parrella NF, Hill AT, Dipnall LM, Loke YJ, Enticott PG, Ford TC. Inhibitory dysfunction and social processing difficulties in autism: A comprehensive narrative review. J Psychiatr Res 2024; 169:113-125. [PMID: 38016393 DOI: 10.1016/j.jpsychires.2023.11.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 09/04/2023] [Accepted: 11/15/2023] [Indexed: 11/30/2023]
Abstract
The primary inhibitory neurotransmitter γ-aminobutyric acid (GABA) has a prominent role in regulating neural development and function, with disruption to GABAergic signalling linked to behavioural phenotypes associated with neurodevelopmental disorders, particularly autism. Such neurochemical disruption, likely resulting from diverse genetic and molecular mechanisms, particularly during early development, can subsequently affect the cellular balance of excitation and inhibition in neuronal circuits, which may account for the social processing difficulties observed in autism and related conditions. This comprehensive narrative review integrates diverse streams of research from several disciplines, including molecular neurobiology, genetics, epigenetics, and systems neuroscience. In so doing it aims to elucidate the relevance of inhibitory dysfunction to autism, with specific focus on social processing difficulties that represent a core feature of this disorder. Many of the social processing difficulties experienced in autism have been linked to higher levels of the excitatory neurotransmitter glutamate and/or lower levels of inhibitory GABA. While current therapeutic options for social difficulties in autism are largely limited to behavioural interventions, this review highlights the psychopharmacological studies that explore the utility of GABA modulation in alleviating such difficulties.
Collapse
Affiliation(s)
| | - Aron T Hill
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia; Department of Psychiatry, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Lillian M Dipnall
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia; Early Life Epigenetics Group, Deakin University, Geelong, Australia
| | - Yuk Jing Loke
- Epigenetics Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Peter G Enticott
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia
| | - Talitha C Ford
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia; Centre for Human Psychopharmacology, Faculty of Health, Arts and Design, Swinburne University of Technology, Melbourne, Victoria, Australia
| |
Collapse
|
10
|
Trofimova I. Anticipatory attractors, functional neurochemistry and "Throw & Catch" mechanisms as illustrations of constructivism. Rev Neurosci 2023; 34:737-762. [PMID: 36584323 DOI: 10.1515/revneuro-2022-0120] [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: 10/01/2022] [Accepted: 12/07/2022] [Indexed: 12/31/2022]
Abstract
This review explores several rarely discussed examples illustrating constructivism principles, generative and selective features of neuronal regulation of behaviour. First, the review highlights Walter Freeman's experiments and mathematical analysis that uncovered the existence of anticipatory attractors, i.e. non-random dynamical patterns in neurodynamics. Since Freeman's work did not extend to neurochemistry, this paper then points to the proposed earlier neurochemical framework summarizing the managerial roles of monoaminergic, cholinergic and opioid receptor systems likely contributing to anticipatory attractors in line with functional constructivism. As a third example, neurochemistry's evidence points to the "Throw & Catch" (T&C) principle in neurodynamics. This principle refers to the pro-active, neurochemically expensive, massive but topical increase of potentials ("Throw") within electrodynamics and neurotransmission in the brain whenever there is an uncertainty in selection of degrees of freedom (DFs). The T&C also underlines the relay-like processes during the selection of DFs. The "Throw" works as an internally generated "flashlight" that, contrarily to the expectations of entropy reduction, increases entropy and variance observed in processes related to orientation and action-formation. The discussed examples highlight the deficiency of structures-oriented projects and excitation-inhibition concepts in neuroscience. The neural regulation of behaviour appears to be a fluid, constructive process, constantly upgrading the choice of behavioural DFs, to ensure the compatibility between the environmental and individual's individuals' needs and capacities.
Collapse
Affiliation(s)
- Irina Trofimova
- Laboratory of Collective Intelligence, Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton L8S 2T6, ON, Canada
| |
Collapse
|
11
|
Nakanishi S, Tamura S, Hirano S, Takahashi J, Kitajima K, Takai Y, Mitsudo T, Togao O, Nakao T, Onitsuka T, Hirano Y. Abnormal phase entrainment of low- and high-gamma-band auditory steady-state responses in schizophrenia. Front Neurosci 2023; 17:1277733. [PMID: 37942136 PMCID: PMC10627971 DOI: 10.3389/fnins.2023.1277733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023] Open
Abstract
Introduction Gamma-band oscillatory deficits have attracted considerable attention as promising biomarkers of schizophrenia (SZ). Notably, a reduced auditory steady-state response (ASSR) in the low gamma band (40 Hz) is widely recognized as a robust finding among SZ patients. However, a comprehensive investigation into the potential utility of the high-gamma-band ASSR in detecting altered neural oscillations in SZ has not yet been conducted. Methods The present study aimed to assess the ASSR using magnetoencephalography (MEG) data obtained during steady-state stimuli at frequencies of 20, 30, 40, and 80 Hz from 23 SZ patients and 21 healthy controls (HCs). To evaluate the ASSR, we examined the evoked power and phase-locking factor (PLF) in the time-frequency domain for both the primary and secondary auditory cortices. Furthermore, we calculated the phase-locking angle (PLA) to examine oscillatory phase lead or delay in SZ patients. Taking advantage of the high spatial resolution of MEG, we also focused on the hemispheric laterality of low- and high-gamma-band ASSR deficits in SZ. Results We found abnormal phase delay in the 40 Hz ASSR within the bilateral auditory cortex of SZ patients. Regarding the 80 Hz ASSR, our investigation identified an aberrant phase lead in the left secondary auditory cortex in SZ, accompanied by reduced evoked power in both auditory cortices. Discussion Given that abnormal phase lead on 80 Hz ASSR exhibited the highest discriminative power between HC and SZ, we propose that the examination of PLA in the 80 Hz ASSR holds significant promise as a robust candidate for identifying neurophysiological endophenotypes associated with SZ. Furthermore, the left-hemisphere phase lead observed in the deficits of 80 Hz PLA aligns with numerous prior studies, which have consistently proposed that SZ is characterized by left-lateralized brain dysfunctions.
Collapse
Affiliation(s)
- Shoichiro Nakanishi
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shunsuke Tamura
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Psychiatry, Division of Clinical Neuroscience, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Shogo Hirano
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Junichi Takahashi
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazutoshi Kitajima
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshifumi Takai
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takako Mitsudo
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Osamu Togao
- Department of Molecular Imaging and Diagnosis, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomohiro Nakao
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshiaki Onitsuka
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- National Hospital Organization Sakakibara Hospital, Mie, Japan
| | - Yoji Hirano
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Psychiatry, Division of Clinical Neuroscience, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
12
|
Asraf K, Zaidan H, Natoor B, Gaisler-Salomon I. Synergistic, long-term effects of glutamate dehydrogenase 1 deficiency and mild stress on cognitive function and mPFC gene and miRNA expression. Transl Psychiatry 2023; 13:248. [PMID: 37419882 DOI: 10.1038/s41398-023-02534-y] [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: 12/11/2022] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 07/09/2023] Open
Abstract
Glutamate abnormalities in the medial prefrontal cortex (mPFC) are associated with cognitive deficits. We previously showed that homozygous deletion of CNS glutamate dehydrogenase 1 (Glud1), a metabolic enzyme critical for glutamate metabolism, leads to schizophrenia-like behavioral abnormalities and increased mPFC glutamate; mice heterozygous for CNS Glud1 deletion (C-Glud1+/- mice) showed no cognitive or molecular abnormalities. Here, we examined the protracted behavioral and molecular effects of mild injection stress on C-Glud1+/- mice. We found spatial and reversal learning deficits, as well as large-scale mPFC transcriptional changes in pathways associated with glutamate and GABA signaling, in stress-exposed C-Glud1+/- mice, but not in their stress-naïve or C-Glud1+/+ littermates. These effects were observed several weeks following stress exposure, and the expression levels of specific glutamatergic and GABAergic genes differentiated between high and low reversal learning performance. An increase in miR203-5p expression immediately following stress may provide a translational regulatory mechanism to account for the delayed effect of stress exposure on cognitive function. Our findings show that chronic glutamate abnormalities interact with acute stress to induce cognitive deficits, and resonate with gene x environment theories of schizophrenia. Stress-exposed C-Glud1+/- mice may model a schizophrenia high-risk population, which is uniquely sensitive to stress-related 'trigger' events.
Collapse
Affiliation(s)
- Kfir Asraf
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa, 3498838, Israel
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa, 3498838, Israel
| | - Hiba Zaidan
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa, 3498838, Israel
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa, 3498838, Israel
| | - Baylasan Natoor
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa, 3498838, Israel
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa, 3498838, Israel
| | - Inna Gaisler-Salomon
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa, 3498838, Israel.
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa, 3498838, Israel.
| |
Collapse
|
13
|
Ummear Raza M, Gautam D, Rorie D, Sivarao DV. Differential Effects of Clozapine and Haloperidol on the 40 Hz Auditory Steady State Response-mediated Phase Resetting in the Prefrontal Cortex of the Female Sprague Dawley Rat. Schizophr Bull 2023; 49:581-591. [PMID: 36691888 PMCID: PMC10154723 DOI: 10.1093/schbul/sbac203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Neural synchrony at gamma frequency (~40 Hz) is important for information processing and is disrupted in schizophrenia. From a drug development perspective, molecules that can improve local gamma synchrony are promising candidates for therapeutic development. HYPOTHESIS Given their differentiated clinical profile, clozapine, and haloperidol may have distinct effects on local gamma synchrony engendered by 40 Hz click trains, the so-called auditory steady-state response (ASSR). STUDY DESIGN Clozapine and haloperidol at doses known to mimic clinically relevant D2 receptor occupancy were evaluated using the ASSR in separate cohorts of female SD rats. RESULTS Clozapine (2.5-10 mg/kg, sc) robustly increased intertrial phase coherence (ITC), across all doses. Evoked response increased but less consistently. Background gamma activity, unrelated to the stimulus, showed a reduction at all doses. Closer scrutiny of the data indicated that clozapine accelerated gamma phase resetting. Thus, clozapine augmented auditory information processing in the gamma frequency range by reducing the background gamma, accelerating the gamma phase resetting and improving phase precision and signal power. Modest improvements in ITC were seen with Haloperidol (0.08 and 0.24 mg/kg, sc) without accelerating phase resetting. Evoked power was unaffected while background gamma was reduced at high doses only, which also caused catalepsy. CONCLUSIONS Using click-train evoked gamma synchrony as an index of local neural network function, we provide a plausible neurophysiological basis for the superior and differentiated profile of clozapine. These observations may provide a neurophysiological template for identifying new drug candidates with a therapeutic potential for treatment-resistant schizophrenia.
Collapse
Affiliation(s)
- Muhammad Ummear Raza
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN
| | - Deepshila Gautam
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN
| | - Dakota Rorie
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN
| | - Digavalli V Sivarao
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN
| |
Collapse
|
14
|
Adraoui FW, Douw L, Martens GJM, Maas DA. Connecting Neurobiological Features with Interregional Dysconnectivity in Social-Cognitive Impairments of Schizophrenia. Int J Mol Sci 2023; 24:ijms24097680. [PMID: 37175387 PMCID: PMC10177877 DOI: 10.3390/ijms24097680] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Schizophrenia (SZ) is a devastating psychiatric disorder affecting about 1% of the world's population. Social-cognitive impairments in SZ prevent positive social interactions and lead to progressive social withdrawal. The neurobiological underpinnings of social-cognitive symptoms remain poorly understood, which hinders the development of novel treatments. At the whole-brain level, an abnormal activation of social brain regions and interregional dysconnectivity within social-cognitive brain networks have been identified as major contributors to these symptoms. At the cellular and subcellular levels, an interplay between oxidative stress, neuroinflammation and N-methyl-D-aspartate receptor hypofunction is thought to underly SZ pathology. However, it is not clear how these molecular processes are linked with interregional dysconnectivity in the genesis of social-cognitive symptoms. Here, we aim to bridge the gap between macroscale (connectivity analyses) and microscale (molecular and cellular mechanistic) knowledge by proposing impaired myelination and the disinhibition of local microcircuits as possible causative biological pathways leading to dysconnectivity and abnormal activity of the social brain. Furthermore, we recommend electroencephalography as a promising translational technique that can foster pre-clinical drug development and discuss attractive drug targets for the treatment of social-cognitive symptoms in SZ.
Collapse
Affiliation(s)
- Florian W Adraoui
- Biotrial, Preclinical Pharmacology Department, 7-9 rue Jean-Louis Bertrand, 35000 Rennes, France
| | - Linda Douw
- Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan, 1081 HZ Amsterdam, The Netherlands
| | - Gerard J M Martens
- Donders Centre for Neuroscience (DCN), Department of Molecular Animal Physiology, Faculty of Science, Donders Institute for Brain, Cognition and Behavior, Radboud University, 6525 GA Nijmegen, The Netherlands
- NeuroDrug Research Ltd., 6525 ED Nijmegen, The Netherlands
| | - Dorien A Maas
- Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
15
|
Herzog LE, Wang L, Yu E, Choi S, Farsi Z, Song BJ, Pan JQ, Sheng M. Mouse mutants in schizophrenia risk genes GRIN2A and AKAP11 show EEG abnormalities in common with schizophrenia patients. Transl Psychiatry 2023; 13:92. [PMID: 36914641 PMCID: PMC10011509 DOI: 10.1038/s41398-023-02393-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/16/2023] Open
Abstract
Schizophrenia is a heterogeneous psychiatric disorder with a strong genetic basis, whose etiology and pathophysiology remain poorly understood. Exome sequencing studies have uncovered rare, loss-of-function variants that greatly increase risk of schizophrenia [1], including loss-of-function mutations in GRIN2A (aka GluN2A or NR2A, encoding the NMDA receptor subunit 2A) and AKAP11 (A-Kinase Anchoring Protein 11). AKAP11 and GRIN2A mutations are also associated with bipolar disorder [2], and epilepsy and developmental delay/intellectual disability [1, 3, 4], respectively. Accessible in both humans and rodents, electroencephalogram (EEG) recordings offer a window into brain activity and display abnormal features in schizophrenia patients. Does loss of Grin2a or Akap11 in mice also result in EEG abnormalities? We monitored EEG in heterozygous and homozygous knockout Grin2a and Akap11 mutant mice compared with their wild-type littermates, at 3- and 6-months of age, across the sleep/wake cycle and during auditory stimulation protocols. Grin2a and Akap11 mutants exhibited increased resting gamma power, attenuated auditory steady-state responses (ASSR) at gamma frequencies, and reduced responses to unexpected auditory stimuli during mismatch negativity (MMN) tests. Sleep spindle density was reduced in a gene dose-dependent manner in Akap11 mutants, whereas Grin2a mutants showed increased sleep spindle density. The EEG phenotypes of Grin2a and Akap11 mutant mice show a variety of abnormal features that overlap considerably with human schizophrenia patients, reflecting systems-level changes caused by Grin2a and Akap11 deficiency. These neurophysiologic findings further substantiate Grin2a and Akap11 mutants as genetic models of schizophrenia and identify potential biomarkers for stratification of schizophrenia patients.
Collapse
Affiliation(s)
- Linnea E Herzog
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Lei Wang
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Eunah Yu
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Soonwook Choi
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Zohreh Farsi
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bryan J Song
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jen Q Pan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Morgan Sheng
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
16
|
Tapias-Espinosa C, Sánchez-González A, Cañete T, Sampedro-Viana D, Castillo-Ruiz MDM, Oliveras I, Tobeña A, Aznar S, Fernández-Teruel A. Decreased activation of parvalbumin interneurons in the medial prefrontal cortex in intact inbred Roman rats with schizophrenia-like reduced sensorimotor gating. Behav Brain Res 2023; 437:114113. [PMID: 36108777 DOI: 10.1016/j.bbr.2022.114113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/24/2022] [Accepted: 09/11/2022] [Indexed: 11/28/2022]
Abstract
Prepulse inhibition (PPI) allows assessing schizophrenia-like sensorimotor gating deficits in rodents. Previous studies indicate that PPI is modulated by the medial prefrontal cortex (mPFC), which is in agreement with our findings showing that PPI differences in the Roman rats are associated with divergences in mPFC activity. Here, we explore whether differences in PPI and mPFC activity in male Roman rats can be explained by (i) differences in the activation (c-Fos) of inhibitory neurons (parvalbumin (PV) interneurons); and/or (ii) reduced excitatory drive (PSD-95) to PV interneurons. Our data show that low PPI in the Roman high-avoidance (RHA) rats is associated with reduced activation of PV interneurons. Moreover, the RHA rats exhibit decreased density of both PV interneurons and PSD-95 puncta on active PV interneurons. These findings point to reduced cortical inhibition as a candidate to explain the schizophrenia-like features observed in RHA rats and support the role of impaired cortical inhibition in schizophrenia.
Collapse
Affiliation(s)
- Carles Tapias-Espinosa
- Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Ana Sánchez-González
- Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Toni Cañete
- Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Daniel Sampedro-Viana
- Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Ignasi Oliveras
- Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Adolf Tobeña
- Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Susana Aznar
- Research Laboratory for Stereology and Neuroscience, Bispebjerg-Frederiksberg Hospital, University Hospital of Copenhagen, Copenhagen, Denmark; Copenhagen Center for Translational Research, Bispebjerg-Frederiksberg Hospital, University Hospital of Copenhagen, Copenhagen, Denmark.
| | - Alberto Fernández-Teruel
- Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
17
|
Wilde M, Constantin L, Thorne PR, Montgomery JM, Scott EK, Cheyne JE. Auditory processing in rodent models of autism: a systematic review. J Neurodev Disord 2022; 14:48. [PMID: 36042393 PMCID: PMC9429780 DOI: 10.1186/s11689-022-09458-6] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 08/07/2022] [Indexed: 11/19/2022] Open
Abstract
Autism is a complex condition with many traits, including differences in auditory sensitivity. Studies in human autism are plagued by the difficulty of controlling for aetiology, whereas studies in individual rodent models cannot represent the full spectrum of human autism. This systematic review compares results in auditory studies across a wide range of established rodent models of autism to mimic the wide range of aetiologies in the human population. A search was conducted in the PubMed and Web of Science databases to find primary research articles in mouse or rat models of autism which investigate central auditory processing. A total of 88 studies were included. These used non-invasive measures of auditory function, such as auditory brainstem response recordings, cortical event-related potentials, electroencephalography, and behavioural tests, which are translatable to human studies. They also included invasive measures, such as electrophysiology and histology, which shed insight on the origins of the phenotypes found in the non-invasive studies. The most consistent results across these studies were increased latency of the N1 peak of event-related potentials, decreased power and coherence of gamma activity in the auditory cortex, and increased auditory startle responses to high sound levels. Invasive studies indicated loss of subcortical inhibitory neurons, hyperactivity in the lateral superior olive and auditory thalamus, and reduced specificity of responses in the auditory cortex. This review compares the auditory phenotypes across rodent models and highlights those that mimic findings in human studies, providing a framework and avenues for future studies to inform understanding of the auditory system in autism.
Collapse
Affiliation(s)
- Maya Wilde
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Lena Constantin
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Peter R Thorne
- Department of Physiology, Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand.,Section of Audiology, School of Population Health, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Johanna M Montgomery
- Department of Physiology, Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Ethan K Scott
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia.,Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Juliette E Cheyne
- Department of Physiology, Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
18
|
Lin L, Zhang J, Dai X, Xiao N, Ye Q, Chen X. A Moderate Duration of Stress Promotes Behavioral Adaptation and Spatial Memory in Young C57BL/6J Mice. Brain Sci 2022; 12:brainsci12081081. [PMID: 36009144 PMCID: PMC9405600 DOI: 10.3390/brainsci12081081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/05/2022] [Accepted: 08/12/2022] [Indexed: 12/02/2022] Open
Abstract
Stress may serve multiple roles in cerebral functioning, ranging from a highly appropriate behavioral adaptation to a critical risk factor for susceptibility to mood disorder and cognitive impairment. It is well known that E/I (excitation/inhibition) balance is essential for maintaining brain homeostasis. However, it remains largely unknown how GABAergic and Glutamatergic neurons respond to different stressful stimuli and whether the GABAergic-Glutamatergic neuron balance is related to the transition between adaptive and maladaptive behaviors. Here, we subjected 3-month-old mice to chronic mild stress (CMS) for a period of one, two, and four weeks, respectively. The results showed that the two-week CMS procedure produced adaptive effects on behaviors and cognitive performance, with a higher number of GABAergic neuron and VGluT1-positive neurons, increasing the expressions of p-GluN2B, Reelin, and syn-PSD-95 protein in the hippocampus. In contrast, the prolonged behavioral challenge (4 week) imposes a passive coping behavioral strategy and cognitive impairment, decreased the number of GABAergic neuron, hyperactivity of VGluT1-positive neuron, increased the ratio of p-GluN2B, and decreased the expression of Reelin, syn-PSD-95 in the hippocampus. These findings suggest that a moderate duration of stress probably promotes behavioral adaptation and spatial memory by maintaining a GABAergic-Glutamatergic neuron balance and promoting the expression of synaptic plasticity-related proteins in the brain.
Collapse
Affiliation(s)
- Lanyan Lin
- Department of Geriatrics, Fujian Provincial Hospital, 134 Dongjie Road, Fuzhou 350001, China
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou 350005, China
| | - Jing Zhang
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou 350005, China
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China
| | - Xiaoman Dai
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou 350005, China
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China
| | - Nai’an Xiao
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou 350005, China
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China
| | - Qinyong Ye
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou 350005, China
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China
| | - Xiaochun Chen
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou 350005, China
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China
- Correspondence: ; Tel.: +86-591-8333-3995; Fax: +86-591-8337-0393
| |
Collapse
|
19
|
Antón-Toro LF, Bruña R, Del Cerro-León A, Shpakivska D, Mateos-Gordo P, Porras-Truque C, García-Gómez R, Maestú F, García-Moreno LM. Electrophysiological resting-state hyperconnectivity and poorer behavioural regulation as predisposing profiles of adolescent binge drinking. Addict Biol 2022; 27:e13199. [PMID: 35754100 PMCID: PMC9286401 DOI: 10.1111/adb.13199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/29/2022] [Accepted: 06/02/2022] [Indexed: 11/30/2022]
Abstract
Adolescent Binge Drinking (BD) has become an increasing health and social concern, with detrimental consequences for brain development and functional integrity. However, research on neurophysiological and neuropsychological traits predisposing to BD are limited at this time. In this work, we conducted a 2‐year longitudinal magnetoencephalography (MEG) study over a cohort of initially alcohol‐naïve adolescents with the purpose of exploring anomalies in resting‐state electrophysiological networks, impulsivity, sensation‐seeking, and dysexecutive behaviour able to predict future BD patterns. In a sample of 67 alcohol‐naïve adolescents (age = 14.5 ± 0.9), we measured resting‐state activity using MEG. Additionally, we evaluated their neuropsychological traits using self‐report ecological scales (BIS‐11, SSS‐V, BDEFS, BRIEF‐SR and DEX). In a second evaluation, 2 years later, we measured participant's alcohol consumption, sub‐dividing the original sample in two groups: future binge drinkers (22 individuals, age 14.6 ± 0.8; eight females) and future light/no drinkers (17 individuals, age 14.5 ± 0.8; eight females). Then, we searched for differences predating alcohol BD intake. We found abnormalities in MEG resting state, in a form of gamma band hyperconnectivity, in those adolescents who transitioned into BD years later. Furthermore, they showed higher impulsivity, dysexecutive behaviours and sensation seeking, positively correlated with functional connectivity (FC). Sensation seeking and impulsivity mainly predicted BD severity in the future, while the relationship between dysexecutive trait and FC with future BD was mediated by sensation seeking. These findings shed light to electrophysiological and neuropsychological traits of vulnerability towards alcohol consumption. We hypothesise that these differences may rely on divergent neurobiological development of inhibitory neurotransmission pathways and executive prefrontal circuits.
Collapse
Affiliation(s)
- Luis F Antón-Toro
- Department of Experimental Psychology, Complutense University of Madrid (UCM), Madrid, Spain
| | - Ricardo Bruña
- Department of Radiology, Complutense University of Madrid (UCM), Madrid, Spain
| | - Alberto Del Cerro-León
- Department of Experimental Psychology, Complutense University of Madrid (UCM), Madrid, Spain
| | - Danylyna Shpakivska
- Department of Experimental Psychology, Complutense University of Madrid (UCM), Madrid, Spain
| | - Patricia Mateos-Gordo
- Department of Psychobiology and Methodology in Behavioral Sciences, Complutense University of Madrid (UCM), Madrid, Spain
| | - Claudia Porras-Truque
- Department of Psychobiology and Methodology in Behavioral Sciences, Complutense University of Madrid (UCM), Madrid, Spain
| | - Raquel García-Gómez
- Department of Psychobiology and Methodology in Behavioral Sciences, Complutense University of Madrid (UCM), Madrid, Spain
| | - Fernando Maestú
- Department of Experimental Psychology, Complutense University of Madrid (UCM), Madrid, Spain
| | - Luis Miguel García-Moreno
- Department of Psychobiology and Methodology in Behavioral Sciences, Complutense University of Madrid (UCM), Madrid, Spain
| |
Collapse
|
20
|
Keeping Excitation-Inhibition Ratio in Balance. Int J Mol Sci 2022; 23:ijms23105746. [PMID: 35628556 PMCID: PMC9145842 DOI: 10.3390/ijms23105746] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 11/17/2022] Open
Abstract
Unrelated genetic mutations can lead to convergent manifestations of neurological disorders with similar behavioral phenotypes. Experimental data frequently show a lack of dramatic changes in neuroanatomy, indicating that the key cause of symptoms might arise from impairment in the communication between neurons. A transient imbalance between excitatory (glutamatergic) and inhibitory (GABAergic) synaptic transmission (the E/I balance) during early development is generally considered to underlie the development of several neurological disorders in adults. However, the E/I ratio is a multidimensional variable. Synaptic contacts are highly dynamic and the actual strength of synaptic projections is determined from the balance between synaptogenesis and synaptic elimination. During development, relatively slow postsynaptic receptors are replaced by fast ones that allow for fast stimulus-locked excitation/inhibition. Using the binomial model of synaptic transmission allows for the reassessing of experimental data from different mouse models, showing that a transient E/I shift is frequently counterbalanced by additional pre- and/or postsynaptic changes. Such changes—for instance, the slowing down of postsynaptic currents by means of immature postsynaptic receptors—stabilize the average synaptic strength, but impair the timing of information flow. Compensatory processes and/or astrocytic signaling may represent possible targets for medical treatments of different disorders directed to rescue the proper information processing.
Collapse
|
21
|
Keeping the Balance: GABAB Receptors in the Developing Brain and Beyond. Brain Sci 2022; 12:brainsci12040419. [PMID: 35447949 PMCID: PMC9031223 DOI: 10.3390/brainsci12040419] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 12/16/2022] Open
Abstract
The main neurotransmitter in the brain responsible for the inhibition of neuronal activity is γ-aminobutyric acid (GABA). It plays a crucial role in circuit formation during development, both via its primary effects as a neurotransmitter and also as a trophic factor. The GABAB receptors (GABABRs) are G protein-coupled metabotropic receptors; on one hand, they can influence proliferation and migration; and, on the other, they can inhibit cells by modulating the function of K+ and Ca2+ channels, doing so on a slower time scale and with a longer-lasting effect compared to ionotropic GABAA receptors. GABABRs are expressed pre- and post-synaptically, at both glutamatergic and GABAergic terminals, thus being able to shape neuronal activity, plasticity, and the balance between excitatory and inhibitory synaptic transmission in response to varying levels of extracellular GABA concentration. Furthermore, given their subunit composition and their ability to form complexes with several associated proteins, GABABRs display heterogeneity with regard to their function, which makes them a promising target for pharmacological interventions. This review will describe (i) the latest results concerning GABABRs/GABABR-complex structures, their function, and the developmental time course of their appearance and functional integration in the brain, (ii) their involvement in manifestation of various pathophysiological conditions, and (iii) the current status of preclinical and clinical studies involving GABABR-targeting drugs.
Collapse
|
22
|
Janz P, Nicolas MJ, Redondo RL, Valencia M.
GABA
B
R
activation partially normalizes acute
NMDAR
hypofunction oscillatory abnormalities but fails to rescue sensory processing deficits. J Neurochem 2022; 161:417-434. [DOI: 10.1111/jnc.15602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/21/2022] [Accepted: 02/12/2022] [Indexed: 12/01/2022]
Affiliation(s)
- Philipp Janz
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann‐La Roche Ltd, Grenzacherstrasse 124, 4070 Basel Switzerland
| | - Maria Jesus Nicolas
- Universidad de Navarra, CIMA, Program of Neuroscience, 31080 Pamplona Spain
- IdiSNA Navarra Institute for Health Research, 31080 Pamplona Spain
| | - Roger L. Redondo
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann‐La Roche Ltd, Grenzacherstrasse 124, 4070 Basel Switzerland
| | - Miguel Valencia
- Universidad de Navarra, CIMA, Program of Neuroscience, 31080 Pamplona Spain
- IdiSNA Navarra Institute for Health Research, 31080 Pamplona Spain
| |
Collapse
|
23
|
Jiang S, Xiao L, Sun Y, He M, Gao C, Zhu C, Chang H, Ding J, Li W, Wang Y, Sun T, Wang F. The GABAB receptor agonist STX209 reverses the autism‑like behaviour in an animal model of autism induced by prenatal exposure to valproic acid. Mol Med Rep 2022; 25:154. [PMID: 35244195 PMCID: PMC8941376 DOI: 10.3892/mmr.2022.12670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/16/2022] [Indexed: 11/06/2022] Open
Abstract
Autism spectrum disorder (ASD) is a lifelong neurodevelopmental condition characterized by impaired social interaction, compromised communication, and restrictive or stereotyped behaviours and interests. Due to the complex pathophysiology of ASD, there are currently no available medical therapies for improving the associated social deficits. Consequently, the present study investigated the effects of STX209, a selective γ‑aminobutyric acid type B receptor (GABABR2) agonist, on an environmental rodent model of autism. The mouse model of autism induced by prenatal exposure to valproic acid (VPA) was used to assess the therapeutic potential of STX209 on autism‑like behaviour in the present study. This study investigated the effects of STX209 on VPA model mice via behavioral testing and revealed a significant reversal of core/associated autism‑like behavior, including sociability and preference for social novelty, novelty recognition, locomotion and exploration activity and marble‑burying deficit. This may be associated with STX209 correcting dendritic arborization, spine density and GABABR2 expression in hippocampus of VPA model mice. However, expression of glutamic acid decarboxylase 65/67 in the hippocampus were not altered by STX209. The present results demonstrated that STX209 administration ameliorated autism‑like symptoms in mice exposed to VPA prenatally, suggesting that autism‑like symptoms in children with a history of prenatal VPA exposure may also benefit from treatment with the GABABR2 agonist STX209.
Collapse
Affiliation(s)
- Shucai Jiang
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Lifei Xiao
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yu Sun
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Maotao He
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Caibin Gao
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Changliang Zhu
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Haigang Chang
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Jiangwei Ding
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Wenchao Li
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yangyang Wang
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Tao Sun
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Feng Wang
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
24
|
Jiang S, He M, Xiao L, Sun Y, Ding J, Li W, Guo B, Wang L, Wang Y, Gao C, Sun T, Wang F. Prenatal GABAB Receptor Agonist Administration Corrects the Inheritance of Autism-Like Core Behaviors in Offspring of Mice Prenatally Exposed to Valproic Acid. Front Psychiatry 2022; 13:835993. [PMID: 35492716 PMCID: PMC9051083 DOI: 10.3389/fpsyt.2022.835993] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/07/2022] [Indexed: 12/21/2022] Open
Abstract
This study was performed to evaluate the effects of prenatal baclofen (a GABAB receptor agonist) treatment on the inheritance of autism-like behaviors in valproic acid (VPA)-exposed mice. VPA model mice (first generation, F1) that were prenatally exposed to VPA exhibited robust core autism-like behaviors, and we found that oral administration of baclofen to F1 mice corrected their autism-like behavioral phenotypes at an early age. Based on a previous epigenetics study, we mated the F1 male offspring with litter females to produce the second generation (F2). The F2 male mice showed obvious inheritance of autism-like phenotypes from F1 mice, implying the heritability of autism symptoms in patients with prenatal VPA exposure. Furthermore, we found prenatal baclofen administration was associated with beneficial effects on the autism-like phenotype in F2 male mice. This may have involved corrections in the density of total/mature dendritic spines in the hippocampus (HC) and medial prefrontal cortex (mPFC), normalizing synaptic plasticity. In this research, GABAB receptor agonist administration corrected the core autism-like behaviors of F1 mice and protected against the inheritance of neurodevelopmental disorders in the offspring of F1 mice, suggesting the potential of early intervention with GABAB receptor agonists in the treatment of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Shucai Jiang
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Maotao He
- School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Lifei Xiao
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Yu Sun
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Jiangwei Ding
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Wenchao Li
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Baorui Guo
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Lei Wang
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Yangyang Wang
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Caibin Gao
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Tao Sun
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Feng Wang
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China.,The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
25
|
Anderson EM, Loke S, Wrucke B, Engelhardt A, Demis S, O'Reilly K, Hess E, Wickman K, Hearing MC. Suppression of pyramidal neuron G protein-gated inwardly rectifying K+ channel signaling impairs prelimbic cortical function and underlies stress-induced deficits in cognitive flexibility in male, but not female, mice. Neuropsychopharmacology 2021; 46:2158-2169. [PMID: 34158613 PMCID: PMC8505646 DOI: 10.1038/s41386-021-01063-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/25/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022]
Abstract
Imbalance in prefrontal cortical (PFC) pyramidal neuron excitation:inhibition is thought to underlie symptomologies shared across stress-related disorders and neuropsychiatric disease, including dysregulation of emotion and cognitive function. G protein-gated inwardly rectifying K+ (GIRK/Kir3) channels mediate excitability of medial PFC pyramidal neurons, however, the functional role of these channels in mPFC-dependent regulation of affect, cognition, and cortical dynamics is unknown. We used a viral-cre approach in male and female mice harboring a "floxed" version of the kcnj3 (Girk1) gene, to disrupt GIRK1-containing channel expression in pyramidal neurons within the prelimbic cortex (PrL). In males, loss of pyramidal GIRK1-dependent signaling differentially impacted measures of affect and impaired working memory and cognitive flexibility. Unexpectedly, ablation of PrL GIRK1-dependent signaling did not impact affect or cognition in female mice. Additional studies used a model of chronic unpredictable stress (CUS) to determine the impact on PrL GIRK-dependent signaling and cognitive function. CUS exposure in male mice produced deficits in cognition that paralleled a reduction in PrL pyramidal GIRK-dependent signaling akin to viral approaches whereas CUS exposure in female mice did not alter cognitive flexibility performance. Stress-induced behavioral deficits in male mice were rescued by systemic injection of a novel, GIRK1-selective agonist, ML297. In conclusion, GIRK1-dependent signaling in male mice, but not females, is critical for maintaining optimal PrL function and behavioral control. Disruption of this inhibition may underlie stress-related dysfunction of the PrL and represent a therapeutic target for treating stress-induced deficits in affect regulation and impaired cognition that reduce quality of life.
Collapse
Affiliation(s)
- Eden M Anderson
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Steven Loke
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Benjamin Wrucke
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Annabel Engelhardt
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Skyler Demis
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Kevin O'Reilly
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Evan Hess
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Matthew C Hearing
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA.
| |
Collapse
|
26
|
Cash RFH, Udupa K, Gunraj CA, Mazzella F, Daskalakis ZJ, Wong AHC, Kennedy JL, Chen R. Influence of BDNF Val66Met polymorphism on excitatory-inhibitory balance and plasticity in human motor cortex. Clin Neurophysiol 2021; 132:2827-2839. [PMID: 34592560 DOI: 10.1016/j.clinph.2021.07.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/30/2021] [Accepted: 07/27/2021] [Indexed: 01/23/2023]
Abstract
OBJECTIVE While previous studies showed that the single nucleotide polymorphism (Val66Met) of brain-derived neurotrophic factor (BDNF) can impact neuroplasticity, the influence of BDNF genotype on cortical circuitry and relationship to neuroplasticity remain relatively unexplored in human. METHODS Using individualised transcranial magnetic stimulation (TMS) parameters, we explored the influence of the BDNF Val66Met polymorphism on excitatory and inhibitory neural circuitry, its relation to I-wave TMS (ITMS) plasticity and effect on the excitatory/inhibitory (E/I) balance in 18 healthy individuals. RESULTS Excitatory and inhibitory indexes of neurotransmission were reduced in Met allele carriers. An E/I balance was evident, which was influenced by BDNF with higher E/I ratios in Val/Val homozygotes. Both long-term potentiation (LTP-) and depression (LTD-) like ITMS plasticity were greater in Val/Val homozygotes. LTP- but not LTD-like effects were restored in Met allele carriers by increasing stimulus intensity to compensate for reduced excitatory transmission. CONCLUSIONS The influence of BDNF genotype may extend beyond neuroplasticity to neurotransmission. The E/I balance was evident in human motor cortex, modulated by BDNF and measurable using TMS. Given the limited sample, these preliminary findings warrant further investigation. SIGNIFICANCE These novel findings suggest a broader role of BDNF genotype on neurocircuitry in human motor cortex.
Collapse
Affiliation(s)
- R F H Cash
- Division of Neurology, Department of Medicine, University of Toronto and Krembil Brain Institute, Toronto, Ontario, Canada; Melbourne Neuropsychiatry Centre, The University of Melbourne, Victoria 3010, Australia; Department of Biomedical Engineering, The University of Melbourne, Victoria 3010, Australia.
| | - K Udupa
- Division of Neurology, Department of Medicine, University of Toronto and Krembil Brain Institute, Toronto, Ontario, Canada; Dept of Neurophysiology, NIMHANS, Bengaluru, India
| | - C A Gunraj
- Division of Neurology, Department of Medicine, University of Toronto and Krembil Brain Institute, Toronto, Ontario, Canada
| | - F Mazzella
- Division of Neurology, Department of Medicine, University of Toronto and Krembil Brain Institute, Toronto, Ontario, Canada
| | - Z J Daskalakis
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, and Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, UC San Diego Health, San Diego, CA 92093, USA
| | - A H C Wong
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, and Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - J L Kennedy
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, and Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - R Chen
- Division of Neurology, Department of Medicine, University of Toronto and Krembil Brain Institute, Toronto, Ontario, Canada
| |
Collapse
|
27
|
Möhrle D, Wang W, Whitehead SN, Schmid S. GABA B Receptor Agonist R-Baclofen Reverses Altered Auditory Reactivity and Filtering in the Cntnap2 Knock-Out Rat. Front Integr Neurosci 2021; 15:710593. [PMID: 34489651 PMCID: PMC8417788 DOI: 10.3389/fnint.2021.710593] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 07/27/2021] [Indexed: 12/23/2022] Open
Abstract
Altered sensory information processing, and auditory processing, in particular, is a common impairment in individuals with autism spectrum disorder (ASD). One prominent hypothesis for the etiology of ASD is an imbalance between neuronal excitation and inhibition. The selective GABAB receptor agonist R-Baclofen has been shown previously to improve social deficits and repetitive behaviors in several mouse models for neurodevelopmental disorders including ASD, and its formulation Arbaclofen has been shown to ameliorate social avoidance symptoms in some individuals with ASD. The present study investigated whether R-Baclofen can remediate ASD-related altered sensory processing reliant on excitation/inhibition imbalance in the auditory brainstem. To assess a possible excitation/inhibition imbalance in the startle-mediating brainstem underlying ASD-like auditory-evoked behaviors, we detected and quantified brain amino acid levels in the nucleus reticularis pontis caudalis (PnC) of rats with a homozygous loss-of-function mutation in the ASD-linked gene Contactin-associated protein-like 2 (Cntnap2) and their wildtype (WT) littermates using Matrix-Assisted Laser Desorption Ionization Mass Spectrometry (MALDI MS). Abnormal behavioral read-outs of brainstem auditory signaling in Cntnap2 KO rats were accompanied by increased levels of GABA, glutamate, and glutamine in the PnC. We then compared the effect of R-Baclofen on behavioral read-outs of brainstem auditory signaling in Cntnap2 KO and WT rats. Auditory reactivity, sensory filtering, and sensorimotor gating were tested in form of acoustic startle response input-output functions, short-term habituation, and prepulse inhibition before and after acute administration of R-Baclofen (0.75, 1.5, and 3 mg/kg). Systemic R-Baclofen treatment improved disruptions in sensory filtering in Cntnap2 KO rats and suppressed exaggerated auditory startle responses, in particular to moderately loud sounds. Lower ASR thresholds in Cntnap2 KO rats were increased in a dose-dependent fashion, with the two higher doses bringing thresholds close to controls, whereas shorter ASR peak latencies at the threshold were further exacerbated. Impaired prepulse inhibition increased across various acoustic prepulse conditions after administration of R-Baclofen in Cntnap2 KO rats, whereas R-Baclofen did not affect prepulse inhibition in WT rats. Our findings suggest that GABAB receptor agonists may be useful for pharmacologically targeting multiple aspects of sensory processing disruptions involving neuronal excitation/inhibition imbalances in ASD.
Collapse
Affiliation(s)
- Dorit Möhrle
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Wenxuan Wang
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Shawn N Whitehead
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Susanne Schmid
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
28
|
Abstract
GABAB receptors are implicated in numerous central nervous system-based behaviours and mechanisms, including cognitive processing in preclinical animal models. Homeostatic changes in the expression and function of these receptors across brain structures have been found to affect cognitive processing. Numerous preclinical studies have focused on the role of GABAB receptors in learning, memory and cognition per se with some interesting, although sometimes contradictory, findings. The majority of the existing clinical literature focuses on alterations in GABAB receptor function in conditions and disorders whose main symptomatology includes deficits in cognitive processing. The aim of this chapter is to delineate the role of GABAB receptors in cognitive processes in health and disease of animal models and human clinical populations. More specifically, this review aims to present literature on the role of GABAB receptors in animal models with cognitive deficits, especially those of learning and memory. Further, it aims to capture the progress and advances of research studies on the effects of GABAB receptor compounds in neurodevelopmental and neurodegenerative conditions with cognitive dysfunctions. The neurodevelopmental conditions covered include autism spectrum disorders, fragile X syndrome and Down's syndrome and the neurodegenerative conditions discussed are Alzheimer's disease, epilepsy and autoimmune anti-GABAB encephalitis. Although some findings are contradictory, results indicate a possible therapeutic role of GABAB receptor compounds for the treatment of cognitive dysfunction and learning/memory impairments for some of these conditions, especially in neurodegeneration. Moreover, future research efforts should aim to develop selective GABAB receptor compounds with minimal, if any, side effects.
Collapse
|
29
|
Kadriu B, Musazzi L, Johnston JN, Kalynchuk LE, Caruncho HJ, Popoli M, Zarate CA. Positive AMPA receptor modulation in the treatment of neuropsychiatric disorders: A long and winding road. Drug Discov Today 2021; 26:2816-2838. [PMID: 34358693 DOI: 10.1016/j.drudis.2021.07.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/12/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022]
Abstract
Glutamatergic transmission is widely implicated in neuropsychiatric disorders, and the discovery that ketamine elicits rapid-acting antidepressant effects by modulating α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) signaling has spurred a resurgence of interest in the field. This review explores agents in various stages of development for neuropsychiatric disorders that positively modulate AMPARs, both directly and indirectly. Despite promising preclinical research, few direct and indirect AMPAR positive modulators have progressed past early clinical development. Challenges such as low potency have created barriers to effective implementation. Nevertheless, the functional complexity of AMPARs sets them apart from other drug targets and allows for specificity in drug discovery. Additional effective treatments for neuropsychiatric disorders that work through positive AMPAR modulation may eventually be developed.
Collapse
Affiliation(s)
- Bashkim Kadriu
- Experimental Therapeutics & Pathophysiology Branch, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Laura Musazzi
- School of Medicine and Surgery, University of Milano-Bicocca, Italy
| | - Jenessa N Johnston
- Experimental Therapeutics & Pathophysiology Branch, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Lisa E Kalynchuk
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Hector J Caruncho
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Maurizio Popoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Italy
| | - Carlos A Zarate
- Experimental Therapeutics & Pathophysiology Branch, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
30
|
McCracken JT, Anagnostou E, Arango C, Dawson G, Farchione T, Mantua V, McPartland J, Murphy D, Pandina G, Veenstra-VanderWeele J. Drug development for Autism Spectrum Disorder (ASD): Progress, challenges, and future directions. Eur Neuropsychopharmacol 2021; 48:3-31. [PMID: 34158222 PMCID: PMC10062405 DOI: 10.1016/j.euroneuro.2021.05.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022]
Abstract
In 2017, facing lack of progress and failures encountered in targeted drug development for Autism Spectrum Disorder (ASD) and related neurodevelopmental disorders, the ISCTM with the ECNP created the ASD Working Group charged to identify barriers to progress and recommending research strategies for the field to gain traction. Working Group international academic, regulatory and industry representatives held multiple in-person meetings, teleconferences, and subgroup communications to gather a wide range of perspectives on lessons learned from extant studies, current challenges, and paths for fundamental advances in ASD therapeutics. This overview delineates the barriers identified, and outlines major goals for next generation biomedical intervention development in ASD. Current challenges for ASD research are many: heterogeneity, lack of validated biomarkers, need for improved endpoints, prioritizing molecular targets, comorbidities, and more. The Working Group emphasized cautious but unwavering optimism for therapeutic progress for ASD core features given advances in the basic neuroscience of ASD and related disorders. Leveraging genetic data, intermediate phenotypes, digital phenotyping, big database discovery, refined endpoints, and earlier intervention, the prospects for breakthrough treatments are substantial. Recommendations include new priorities for expanded research funding to overcome challenges in translational clinical ASD therapeutic research.
Collapse
Affiliation(s)
- James T McCracken
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024, United States.
| | | | - Celso Arango
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Univesitario Gregorio Maranon, and School of Medicine, Universidad Complutense de Madrid, CIBERSAM, Madrid, Spain
| | - Geraldine Dawson
- Duke University Medical Center, Durham, North Carolina, United States
| | - Tiffany Farchione
- Food and Drug Administration, Silver Spring, Maryland, United States
| | - Valentina Mantua
- Food and Drug Administration, Silver Spring, Maryland, United States
| | | | - Declan Murphy
- Institute of Psychiatry, Psychology and Neuroscience, King's College De Crespigny Park, Denmark Hill, London SE5 8AF, United Kingdom
| | - Gahan Pandina
- Neuroscience Therapeutic Area, Janssen Research & Development, Pennington, New Jersey, United States
| | | |
Collapse
|
31
|
Côté V, Knoth IS, Agbogba K, Vannasing P, Côté L, Major P, Michaud JL, Barlaam F, Lippé S. Differential auditory brain response abnormalities in two intellectual disability conditions: SYNGAP1 mutations and Down syndrome. Clin Neurophysiol 2021; 132:1802-1812. [PMID: 34130248 DOI: 10.1016/j.clinph.2021.03.054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 03/06/2021] [Accepted: 03/31/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Altered sensory processing is common in intellectual disability (ID). Here, we study electroencephalographic responses to auditory stimulation in human subjects presenting a rare condition (mutations in SYNGAP1) which causes ID, epilepsy and autism. METHODS Auditory evoked potentials, time-frequency and inter-trial coherence analyses were used to compare subjects with SYNGAP1 mutations with Down syndrome (DS) and neurotypical (NT) participants (N = 61 ranging from three to 19 years of age). RESULTS Altered synchronization in the brain responses to sound were found in both ID groups. The SYNGAP1 mutations group showed less phase-locking in early time windows and lower frequency bands compared to NT, and in later time windows compared to NT and DS. Time-frequency analysis showed more power in beta-gamma in the SYNGAP1 group compared to NT participants. CONCLUSIONS This study indicated reduced synchronization as well as more high frequencies power in SYNGAP1 mutations, while maintained synchronization was found in the DS group. These results might reflect dysfunctional sensory information processing caused by excitation/inhibition imbalance, or an imperfect compensatory mechanism in SYNGAP1 mutations individuals. SIGNIFICANCE Our study is the first to reveal brain response abnormalities in auditory sensory processing in SYNGAP1 mutations individuals, that are distinct from DS, another ID condition.
Collapse
Affiliation(s)
- Valérie Côté
- Department of Psychology, University of Montreal, Montreal, Québec, Canada; CHU Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - Inga S Knoth
- CHU Sainte-Justine Research Center, Montreal, Quebec, Canada
| | | | | | - Lucie Côté
- CHU Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - Philippe Major
- CHU Sainte-Justine Research Center, Montreal, Quebec, Canada; Department of Pediatrics and Neurosciences, University of Montreal, Montreal, Quebec, Canada
| | - Jacques L Michaud
- CHU Sainte-Justine Research Center, Montreal, Quebec, Canada; Department of Pediatrics and Neurosciences, University of Montreal, Montreal, Quebec, Canada
| | - Fanny Barlaam
- CHU Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - Sarah Lippé
- Department of Psychology, University of Montreal, Montreal, Québec, Canada; CHU Sainte-Justine Research Center, Montreal, Quebec, Canada.
| |
Collapse
|
32
|
Anderson EM, Demis S, D’Acquisto H, Engelhardt A, Hearing M. The Role of Parvalbumin Interneuron GIRK Signaling in the Regulation of Affect and Cognition in Male and Female Mice. Front Behav Neurosci 2021; 15:621751. [PMID: 33841107 PMCID: PMC8032990 DOI: 10.3389/fnbeh.2021.621751] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/19/2021] [Indexed: 11/13/2022] Open
Abstract
Pathological impairments in the regulation of affect (i.e., emotion) and flexible decision-making are commonly observed across numerous neuropsychiatric disorders and are thought to reflect dysfunction of cortical and subcortical circuits that arise in part from imbalances in excitation and inhibition within these structures. Disruptions in GABA transmission, in particular, that from parvalbumin-expressing interneurons (PVI), has been highlighted as a likely mechanism by which this imbalance arises, as they regulate excitation and synchronization of principle output neurons. G protein-gated inwardly rectifying potassium ion (GIRK/Kir3) channels are known to modulate excitability and output of pyramidal neurons in areas like the medial prefrontal cortex and hippocampus; however, the role GIRK plays in PVI excitability and behavior is unknown. Male and female mice lacking GIRK1 in PVI (Girk1flox/flox:PVcre) and expressing td-tomato in PVI (Girk1flox/flox:PVCre:PVtdtom) exhibited increased open arm time in the elevated plus-maze, while males showed an increase in immobile episodes during the forced swim test (FST). Loss of GIRK1 did not alter motivated behavior for an appetitive reward or impair overall performance in an operant-based attention set-shifting model of cognitive flexibility; however it did alter types of errors committed during the visual cue test. Unexpectedly, baseline sex differences were also identified in these tasks, with females exhibiting overall poorer performance compared to males and distinct types of errors, highlighting potential differences in task-related problem-solving. Interestingly, reductions in PVI GIRK signaling did not correspond to changes in membrane excitability but did increase action potential (AP) firing at higher current injections in PVI of males, but not females. This is the first investigation on the role that PVI GIRK-signaling has on membrane excitability, AP firing, and their role on affect and cognition together increasing the understanding of PVI cellular mechanisms and function.
Collapse
Affiliation(s)
| | | | | | | | - Matthew Hearing
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, United States
| |
Collapse
|
33
|
Takeda K, Watanabe T, Oyabu K, Tsukamoto S, Oba Y, Nakano T, Kubota K, Katsurabayashi S, Iwasaki K. Valproic acid-exposed astrocytes impair inhibitory synapse formation and function. Sci Rep 2021; 11:23. [PMID: 33420078 PMCID: PMC7794250 DOI: 10.1038/s41598-020-79520-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
Valproic acid (VPA) is widely prescribed to treat epilepsy. Maternal VPA use is, however, clinically restricted because of the severe risk that VPA may cause neurodevelopmental disorders in offspring, such as autism spectrum disorder. Understanding the negative action of VPA may help to prevent VPA-induced neurodevelopmental disorders. Astrocytes play a vital role in neurodevelopment and synapse function; however, the impact of VPA on astrocyte involvement in neurodevelopment and synapse function has not been examined. In this study, we examined whether exposure of cultured astrocytes to VPA alters neuronal morphology and synapse function of co-cultured neurons. We show that synaptic transmission by inhibitory neurons was small because VPA-exposed astrocytes reduced the number of inhibitory synapses. However, synaptic transmission by excitatory neurons and the number of excitatory synapses were normal with VPA-exposed astrocytes. VPA-exposed astrocytes did not affect the morphology of inhibitory neurons. These data indicate that VPA-exposed astrocytes impair synaptogenesis specifically of inhibitory neurons. Our results indicate that maternal use of VPA would affect not only neurons but also astrocytes and would result in perturbed astrocyte-mediated neurodevelopment.
Collapse
Affiliation(s)
- Kotomi Takeda
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Takuya Watanabe
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180, Japan. .,A.I.G. Collaborative Research Institute for Aging and Brain Sciences, Fukuoka University, Fukuoka, 814-0180, Japan.
| | - Kohei Oyabu
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Shuntaro Tsukamoto
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Yuki Oba
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Takafumi Nakano
- Department of Pharmaceutical and Health Care Management, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Kaori Kubota
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180, Japan.,A.I.G. Collaborative Research Institute for Aging and Brain Sciences, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Shutaro Katsurabayashi
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Katsunori Iwasaki
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180, Japan.,A.I.G. Collaborative Research Institute for Aging and Brain Sciences, Fukuoka University, Fukuoka, 814-0180, Japan
| |
Collapse
|
34
|
Ballard ED, Zarate CA. The role of dissociation in ketamine's antidepressant effects. Nat Commun 2020; 11:6431. [PMID: 33353946 PMCID: PMC7755908 DOI: 10.1038/s41467-020-20190-4] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 11/17/2020] [Indexed: 02/07/2023] Open
Abstract
Ketamine produces immediate antidepressant effects and has inspired research into next-generation treatments. Ketamine also has short term dissociative effects, in which individuals report altered consciousness and perceptions of themselves and their environment. However, whether ketamine's dissociative side effects are necessary for its antidepressant effects remains unclear. This perspective examines the relationship between dissociative effects and acute and longer-lasting antidepressant response to ketamine and other N-methyl-D-aspartate (NMDA) receptor antagonists. Presently, the literature does not support the conclusion that dissociation is necessary for antidepressant response to ketamine. However, further work is needed to explore the relationship between dissociation and antidepressant response at the molecular, biomarker, and psychological levels.
Collapse
Affiliation(s)
- Elizabeth D Ballard
- Section on the Neurobiology and Treatment of Mood Disorders, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Carlos A Zarate
- Section on the Neurobiology and Treatment of Mood Disorders, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
35
|
Shaw AD, Muthukumaraswamy SD, Saxena N, Sumner RL, Adams NE, Moran RJ, Singh KD. Generative modelling of the thalamo-cortical circuit mechanisms underlying the neurophysiological effects of ketamine. Neuroimage 2020; 221:117189. [PMID: 32711064 PMCID: PMC7762824 DOI: 10.1016/j.neuroimage.2020.117189] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/15/2020] [Accepted: 07/19/2020] [Indexed: 11/25/2022] Open
Abstract
Cortical recordings of task-induced oscillations following subanaesthetic ketamine administration demonstrate alterations in amplitude, including increases at high-frequencies (gamma) and reductions at low frequencies (theta, alpha). To investigate the population-level interactions underlying these changes, we implemented a thalamo-cortical model (TCM) capable of recapitulating broadband spectral responses. Compared with an existing cortex-only 4-population model, Bayesian Model Selection preferred the TCM. The model was able to accurately and significantly recapitulate ketamine-induced reductions in alpha amplitude and increases in gamma amplitude. Parameter analysis revealed no change in receptor time-constants but significant increases in select synaptic connectivity with ketamine. Significantly increased connections included both AMPA and NMDA mediated connections from layer 2/3 superficial pyramidal cells to inhibitory interneurons and both GABAA and NMDA mediated within-population gain control of layer 5 pyramidal cells. These results support the use of extended generative models for explaining oscillatory data and provide in silico support for ketamine's ability to alter local coupling mediated by NMDA, AMPA and GABA-A.
Collapse
Affiliation(s)
- Alexander D Shaw
- Cardiff University Brain Research Imaging Centre (CUBRIC), Cardiff University, Maindy Road, Cardiff CF24 4HQ, UK.
| | - Suresh D Muthukumaraswamy
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Neeraj Saxena
- Cardiff University Brain Research Imaging Centre (CUBRIC), Cardiff University, Maindy Road, Cardiff CF24 4HQ, UK; Department of Anaesthetics, Intensive Care and Pain Medicine, Cwm Taf Morgannwg University Health Board, Llantrisant CF72 8XR, UK
| | - Rachael L Sumner
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Natalie E Adams
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Rosalyn J Moran
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE5 8AF, UK
| | - Krish D Singh
- Cardiff University Brain Research Imaging Centre (CUBRIC), Cardiff University, Maindy Road, Cardiff CF24 4HQ, UK
| |
Collapse
|
36
|
Caruso A, Ricceri L, Scattoni ML. Ultrasonic vocalizations as a fundamental tool for early and adult behavioral phenotyping of Autism Spectrum Disorder rodent models. Neurosci Biobehav Rev 2020; 116:31-43. [DOI: 10.1016/j.neubiorev.2020.06.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 05/08/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022]
|
37
|
Javitt DC, Siegel SJ, Spencer KM, Mathalon DH, Hong LE, Martinez A, Ehlers CL, Abbas AI, Teichert T, Lakatos P, Womelsdorf T. A roadmap for development of neuro-oscillations as translational biomarkers for treatment development in neuropsychopharmacology. Neuropsychopharmacology 2020; 45:1411-1422. [PMID: 32375159 PMCID: PMC7360555 DOI: 10.1038/s41386-020-0697-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 03/16/2020] [Accepted: 04/27/2020] [Indexed: 02/08/2023]
Abstract
New treatment development for psychiatric disorders depends critically upon the development of physiological measures that can accurately translate between preclinical animal models and clinical human studies. Such measures can be used both as stratification biomarkers to define pathophysiologically homogeneous patient populations and as target engagement biomarkers to verify similarity of effects across preclinical and clinical intervention. Traditional "time-domain" event-related potentials (ERP) have been used translationally to date but are limited by the significant differences in timing and distribution across rodent, monkey and human studies. By contrast, neuro-oscillatory responses, analyzed within the "time-frequency" domain, are relatively preserved across species permitting more precise translational comparisons. Moreover, neuro-oscillatory responses are increasingly being mapped to local circuit mechanisms and may be useful for investigating effects of both pharmacological and neuromodulatory interventions on excitatory/inhibitory balance. The present paper provides a roadmap for development of neuro-oscillatory responses as translational biomarkers in neuropsychiatric treatment development.
Collapse
Affiliation(s)
- Daniel C Javitt
- Department of Psychiatry, Columbia University Medical Center, New York, NY, 10032, USA.
- Schizophrenia Research Division, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10954, USA.
| | - Steven J Siegel
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Kevin M Spencer
- Research Service, VA Boston Healthcare System, and Dept. of Psychiatry, Harvard Medical School, Boston, MA, 02130, USA
| | - Daniel H Mathalon
- VA San Francisco Healthcare System, University of California, San Francisco, San Francisco, CA, 94121, USA
| | - L Elliot Hong
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Antigona Martinez
- Department of Psychiatry, Columbia University Medical Center, New York, NY, 10032, USA
- Schizophrenia Research Division, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10954, USA
| | - Cindy L Ehlers
- Department of Neuroscience, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Atheir I Abbas
- VA Portland Health Care System, Portland, OR, 97239, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Tobias Teichert
- Departments of Psychiatry and Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Peter Lakatos
- Schizophrenia Research Division, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10954, USA
| | - Thilo Womelsdorf
- Department of Psychology, Vanderbilt University, Nashville, TN, 37203, USA
| |
Collapse
|
38
|
Nardone R, Sebastianelli L, Brigo F, Golaszewski S, Trinka E, Pucks-Faes E, Saltuari L, Versace V. Effects of intrathecal baclofen therapy in subjects with disorders of consciousness: a reappraisal. J Neural Transm (Vienna) 2020; 127:1209-1215. [PMID: 32710152 DOI: 10.1007/s00702-020-02233-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 07/14/2020] [Indexed: 01/16/2023]
Abstract
Baclofen is a structural analogue of gamma-amino-butyric acid (GABA), which reduces spastic hypertonia of striated muscle due to a mechanism of GABAB-ergic inhibition of mono- and polysynaptic reflexes at the spinal level. There are reports of patients with severe disorders of consciousness that presented a substantial improvement following intrathecal baclofen (ITB) administration for severe spasticity. The neural mechanisms underlying the clinical recovery after ITB have not yet been clarified. Baclofen could modulate sleep-wake cycles that may be dysregulated and thus interfere with alertness and awareness. The diminished proprioceptive and nociceptive sensory inputs may relieve thalamo-cortical neural networks involved in maintaining the consciousness of the self and the world. ITB treatment might also promote the recovery of an impaired GABAergic cortical tone, restoring the balance between excitatory and inhibitory cortical activity. Furthermore, glutamatergic synapses are directly or indirectly modulated by GABAB-ergic receptors. Neurophysiological techniques (such as transcranial magnetic stimulation, electroencephalography, or the combination of both) can be helpful to explore the effects of intrathecal or oral baclofen on the modulation of neural cortical circuits in humans with disorders of consciousness.
Collapse
Affiliation(s)
- Raffaele Nardone
- Department of Neurology, Hospital of Merano (SABES-ASDAA), Merano-Meran, Italy. .,Department of Neurology, Christian Doppler Klinik, Paracelsus Medical University, Salzburg, Austria. .,Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Austria. .,Karl Landsteiner Institut für Neurorehabilitation und Raumfahrtneurologie, Salzburg, Austria.
| | - Luca Sebastianelli
- Department of Neurorehabilitation, Hospital of Vipiteno, Vipiteno, Italy.,Research Unit for Neurorehabilitation South Tyrol, Bolzano, Italy
| | - Francesco Brigo
- Department of Neurology, Hospital of Merano (SABES-ASDAA), Merano-Meran, Italy.,Department of Neuroscience, Biomedicine and Movement Science, University of Verona, Verona, Italy
| | - Stefan Golaszewski
- Department of Neurology, Christian Doppler Klinik, Paracelsus Medical University, Salzburg, Austria.,Karl Landsteiner Institut für Neurorehabilitation und Raumfahrtneurologie, Salzburg, Austria
| | - Eugen Trinka
- Department of Neurology, Christian Doppler Klinik, Paracelsus Medical University, Salzburg, Austria.,Centre for Cognitive Neurosciences Salzburg, Salzburg, Austria.,University for Medical Informatics and Health Technology, UMIT, Hall in Tirol, Austria
| | | | - Leopold Saltuari
- Department of Neurorehabilitation, Hospital of Vipiteno, Vipiteno, Italy.,Research Unit for Neurorehabilitation South Tyrol, Bolzano, Italy
| | - Viviana Versace
- Department of Neurology, Hospital of Merano (SABES-ASDAA), Merano-Meran, Italy.,Research Unit for Neurorehabilitation South Tyrol, Bolzano, Italy
| |
Collapse
|
39
|
Honda S, Matsumoto M, Tajinda K, Mihara T. Enhancing Clinical Trials Through Synergistic Gamma Power Analysis. Front Psychiatry 2020; 11:537. [PMID: 32587536 PMCID: PMC7299152 DOI: 10.3389/fpsyt.2020.00537] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/26/2020] [Indexed: 12/14/2022] Open
Abstract
While the etiology of many neuropsychiatric disorders remains unknown, increasing evidence suggests that aberrant sensory processing plays a central role. For this class of disorders, which are characterized by affective, cognitive, and behavioral symptoms, electroencephalography remains the dominant tool for providing insight into the physiological and molecular underpinnings of the disease state and predicting the effectiveness of investigational new drugs. Within the spectrum of electrical activity present in the CNS, high-frequency oscillations in the gamma band are frequently altered in these patient populations. Measurement of gamma oscillation can be further classified into baseline and evoked, each of which offers a specific commentary on disease state. Baseline gamma analysis provides a surrogate of pharmacodynamics and predicting the time course effects of clinical candidate drugs, while alterations in evoked (time-locked) gamma power may serve as a disease biomarker and have utility in assessing patient response to new drugs. Together, these techniques offer complimentary methods of analysis for discrete realms of clinical and translational medicine. In terms of drug development, comprehensive analysis containing aspects of both baseline and evoked gamma oscillations may prove more useful in establishing better workflow and more accurate criteria for the testing of investigational new drugs.
Collapse
Affiliation(s)
- Sokichi Honda
- Neuroscience, La Jolla Laboratory, Astellas Research Institute of America LLC, San Diego, CA, United States
| | - Mitsuyuki Matsumoto
- Neuroscience, La Jolla Laboratory, Astellas Research Institute of America LLC, San Diego, CA, United States
| | - Katsunori Tajinda
- Neuroscience, La Jolla Laboratory, Astellas Research Institute of America LLC, San Diego, CA, United States
| | - Takuma Mihara
- Candidate Discovery Research Labs, DDR, Astellas Pharm Inc., Tsukuba, Japan
| |
Collapse
|
40
|
Davis BA, David F, O’Regan C, Adam MA, Harwood AJ, Crunelli V, Isles AR. Impairments in sensory-motor gating and information processing in a mouse model of Ehmt1 haploinsufficiency. Brain Neurosci Adv 2020; 4:2398212820928647. [PMID: 32954001 PMCID: PMC7479861 DOI: 10.1177/2398212820928647] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 04/30/2020] [Indexed: 12/14/2022] Open
Abstract
Regulators of chromatin dynamics and transcription are increasingly implicated in the aetiology of neurodevelopmental disorders. Haploinsufficiency of EHMT1, encoding a histone methyltransferase, is associated with several neurodevelopmental disorders, including Kleefstra syndrome, developmental delay and autism spectrum disorder. Using a mouse model of Ehmt1 haploinsufficiency (Ehmt1 D6Cre/+), we examined a number of brain and behavioural endophenotypes of relevance to neurodevelopmental disorders. Specifically, we show that Ehmt1 D6Cre/+ mice have deficits in information processing, evidenced by abnormal sensory-motor gating, a complete absence of object recognition memory, and a reduced magnitude of auditory evoked potentials in both paired-pulse inhibition and mismatch negativity. The electrophysiological experiments show that differences in magnitude response to auditory stimulus were associated with marked reductions in total and evoked beta- and gamma-band oscillatory activity, as well as significant reductions in phase synchronisation. The pattern of electrophysiological deficits in Ehmt1 D6Cre/+ matches those seen in control mice following administration of the selective NMDA-R antagonist, ketamine. This, coupled with reduction of Grin1 mRNA expression in Ehmt1 D6Cre/+ hippocampus, suggests that Ehmt1 haploinsufficiency may lead to disruption in NMDA-R. Taken together, these data indicate that reduced Ehmt1 dosage during forebrain development leads to abnormal circuitry formation, which in turn results in profound information processing deficits. Such information processing deficits are likely paramount to our understanding of the cognitive and neurological dysfunctions shared across the neurodevelopmental disorders associated with EHMT1 haploinsufficiency.
Collapse
Affiliation(s)
- Brittany A Davis
- Neuroscience and Mental Health
Research Institute and School of Biosciences, Cardiff University, Cardiff,
UK
| | - François David
- Neuroscience and Mental Health
Research Institute and School of Biosciences, Cardiff University, Cardiff,
UK
| | - Ciara O’Regan
- MRC Centre for Neuropsychiatric
Genetics and Genomics, School of Medicine, Cardiff University, Cardiff,
UK
| | - Manal A Adam
- MRC Centre for Neuropsychiatric
Genetics and Genomics, School of Medicine, Cardiff University, Cardiff,
UK
| | - Adrian J Harwood
- Neuroscience and Mental Health
Research Institute and School of Biosciences, Cardiff University, Cardiff,
UK
| | - Vincenzo Crunelli
- Neuroscience and Mental Health
Research Institute and School of Biosciences, Cardiff University, Cardiff,
UK
| | - Anthony R Isles
- MRC Centre for Neuropsychiatric
Genetics and Genomics, School of Medicine, Cardiff University, Cardiff,
UK
| |
Collapse
|
41
|
Pianp deficiency links GABA B receptor signaling and hippocampal and cerebellar neuronal cell composition to autism-like behavior. Mol Psychiatry 2020; 25:2979-2993. [PMID: 31511635 PMCID: PMC7577901 DOI: 10.1038/s41380-019-0519-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 05/31/2019] [Accepted: 07/18/2019] [Indexed: 12/17/2022]
Abstract
Pianp (also known as Leda-1) is a type I transmembrane protein with preferential expression in the mammalian CNS. Its processing is characterized by proteolytic cleavage by a range of proteases including Adam10, Adam17, MMPs, and the γ-secretase complex. Pianp can interact with Pilrα and the GB1a subunit of the GABAB receptor (GBR) complex. A recent case description of a boy with global developmental delay and homozygous nonsense variant in PIANP supports the hypothesis that PIANP is involved in the control of behavioral traits in mammals. To investigate the physiological functions of Pianp, constitutive, global knockout mice were generated and comprehensively analyzed. Broad assessment did not indicate malformation or malfunction of internal organs. In the brain, however, decreased sizes and altered cellular compositions of the dentate gyrus as well as the cerebellum, including a lower number of cerebellar Purkinje cells, were identified. Functionally, loss of Pianp led to impaired presynaptic GBR-mediated inhibition of glutamate release and altered gene expression in the cortex, hippocampus, amygdala, and hypothalamus including downregulation of Erdr1, a gene linked to autism-like behavior. Behavioral phenotyping revealed that Pianp deficiency leads to context-dependent enhanced anxiety and spatial learning deficits, an altered stress response, severely impaired social interaction, and enhanced repetitive behavior, which all represent characteristic features of an autism spectrum disorder-like phenotype. Altogether, Pianp represents a novel candidate gene involved in autism-like behavior, cerebellar and hippocampal pathology, and GBR signaling.
Collapse
|
42
|
Ferri SL, Pallathra AA, Kim H, Dow HC, Raje P, McMullen M, Bilker WB, Siegel SJ, Abel T, Brodkin ES. Sociability development in mice with cell-specific deletion of the NMDA receptor NR1 subunit gene. GENES BRAIN AND BEHAVIOR 2019; 19:e12624. [PMID: 31721416 DOI: 10.1111/gbb.12624] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 11/06/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022]
Abstract
Social affiliative behavior is an important component of everyday life in many species and is likely to be disrupted in disabling ways in various neurodevelopmental and neuropsychiatric disorders. Therefore, determining the mechanisms involved in these processes is crucial. A link between N-methyl-d-aspartate (NMDA) receptor function and social behaviors has been clearly established. The cell types in which NMDA receptors are critical for social affiliative behavior, however, remain unclear. Here, we use mice carrying a conditional allele of the NMDA R1 subunit to address this question. Mice bearing a floxed NMDAR1 (NR1) allele were crossed with transgenic calcium/calmodulin-dependent kinase IIα (CaMKIIα)-Cre mice or parvalbumin (PV)-Cre mice targeting postnatal excitatory forebrain or PV-expressing interneurons, respectively, and assessed using the three-chambered Social Approach Test. We found that deletion of NR1 in PV-positive interneurons had no effect on social sniffing, but deletion of NR1 in glutamatergic pyramidal cells resulted in a significant increase in social approach behavior, regardless of age or sex. Therefore, forebrain excitatory neurons expressing NR1 play an important role in regulating social affiliative behavior.
Collapse
Affiliation(s)
- Sarah L Ferri
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa
| | - Ashley A Pallathra
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hyong Kim
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Holly C Dow
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Praachi Raje
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mary McMullen
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Warren B Bilker
- Department of Biostatistics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Steven J Siegel
- Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa
| | - Edward S Brodkin
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
43
|
Port RG, Oberman LM, Roberts TPL. Revisiting the excitation/inhibition imbalance hypothesis of ASD through a clinical lens. Br J Radiol 2019; 92:20180944. [PMID: 31124710 PMCID: PMC6732925 DOI: 10.1259/bjr.20180944] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 04/19/2019] [Accepted: 05/21/2019] [Indexed: 12/22/2022] Open
Abstract
Autism spectrum disorder (ASD) currently affects 1 in 59 children, although the aetiology of this disorder remains unknown. Faced with multiple seemingly disparate and noncontiguous neurobiological alterations, Rubenstein and Merzenich hypothesized that imbalances between excitatory and inhibitory neurosignaling (E/I imbalance) underlie ASD. Since this initial statement, there has been a major focus examining this exact topic spanning both clinical and preclinical realms. The purpose of this article is to review the clinical neuroimaging literature surrounding E/I imbalance as an aetiology of ASD. Evidence for E/I imbalance is presented from several complementary clinical techniques including magnetic resonance spectroscopy, magnetoencephalography and transcranial magnetic stimulation. Additionally, two GABAergic potential interventions for ASD, which explicitly attempt to remediate E/I imbalance, are reviewed. The current literature suggests E/I imbalance as a useful framework for discussing the neurobiological etiology of ASD in at least a subset of affected individuals. While not constituting a completely unifying aetiology, E/I imbalance may be relevant as one of several underlying neuropathophysiologies that differentially affect individuals with ASD. Such statements do not diminish the value of the E/I imbalance concept-instead they suggest a possible role for the characterization of E/I imbalance, as well as other underlying neuropathophysiologies, in the biologically-based subtyping of individuals with ASD for potential applications including clinical trial enrichment as well as treatment triage.
Collapse
Affiliation(s)
| | - Lindsay M Oberman
- Center for Neuroscience and Regenerative Medicine, Henry M. Jackson Foundation for the Advancement of Military Medicine, Rockville, Maryland
| | - Timothy PL Roberts
- Department of Radiology, Lurie Family Foundations MEG Imaging Center, Children’s Hospital of Philadelphia, Pennsylvania
| |
Collapse
|
44
|
Nugent AC, Ballard ED, Gould TD, Park LT, Moaddel R, Brutsche NE, Zarate CA. Ketamine has distinct electrophysiological and behavioral effects in depressed and healthy subjects. Mol Psychiatry 2019; 24:1040-1052. [PMID: 29487402 PMCID: PMC6111001 DOI: 10.1038/s41380-018-0028-2] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 07/13/2017] [Accepted: 11/03/2017] [Indexed: 01/19/2023]
Abstract
Ketamine's mechanism of action was assessed using gamma power from magnetoencephalography (MEG) as a proxy measure for homeostatic balance in 35 unmedicated subjects with major depressive disorder (MDD) and 25 healthy controls enrolled in a double-blind, placebo-controlled, randomized cross-over trial of 0.5 mg/kg ketamine. MDD subjects showed significant improvements in depressive symptoms, and healthy control subjects exhibited modest but significant increases in depressive symptoms for up to 1 day after ketamine administration. Both groups showed increased resting gamma power following ketamine. In MDD subjects, gamma power was not associated with the magnitude of the antidepressant effect. However, baseline gamma power was found to moderate the relationship between post-ketamine gamma power and antidepressant response; specifically, higher post-ketamine gamma power was associated with better response in MDD subjects with lower baseline gamma, with an inverted relationship in MDD subjects with higher baseline gamma. This relationship was observed in multiple regions involved in networks hypothesized to be involved in the pathophysiology of MDD. This finding suggests biological subtypes based on the direction of homeostatic dysregulation and has important implications for inferring ketamine's mechanism of action from studies of healthy controls alone.
Collapse
Affiliation(s)
- Allison C Nugent
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Elizabeth D Ballard
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Lawrence T Park
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Ruin Moaddel
- Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - Nancy E Brutsche
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
45
|
Sullivan CR, Mielnik CA, O'Donovan SM, Funk AJ, Bentea E, DePasquale EA, Alganem K, Wen Z, Haroutunian V, Katsel P, Ramsey AJ, Meller J, McCullumsmith RE. Connectivity Analyses of Bioenergetic Changes in Schizophrenia: Identification of Novel Treatments. Mol Neurobiol 2019; 56:4492-4517. [PMID: 30338483 PMCID: PMC7584383 DOI: 10.1007/s12035-018-1390-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/11/2018] [Indexed: 01/21/2023]
Abstract
We utilized a cell-level approach to examine glycolytic pathways in the DLPFC of subjects with schizophrenia (n = 16) and control (n = 16) and found decreased mRNA expression of glycolytic enzymes in pyramidal neurons, but not astrocytes. To replicate these novel bioenergetic findings, we probed independent datasets for bioenergetic targets and found similar abnormalities. Next, we used a novel strategy to build a schizophrenia bioenergetic profile by a tailored application of the Library of Integrated Network-Based Cellular Signatures data portal (iLINCS) and investigated connected cellular pathways, kinases, and transcription factors using Enrichr. Finally, with the goal of identifying drugs capable of "reversing" the bioenergetic schizophrenia signature, we performed a connectivity analysis with iLINCS and identified peroxisome proliferator-activated receptor (PPAR) agonists as promising therapeutic targets. We administered a PPAR agonist to the GluN1 knockdown model of schizophrenia and found it improved long-term memory. Taken together, our findings suggest that tailored bioinformatics approaches, coupled with the LINCS library of transcriptional signatures of chemical and genetic perturbagens, may be employed to identify novel treatment strategies for schizophrenia and related diseases.
Collapse
Affiliation(s)
| | - Catharine A Mielnik
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | | | - Adam J Funk
- Department of Neuroscience, University of Toledo, Toledo, OH, USA
| | - Eduard Bentea
- Neurosciences TA Biology, UCB BioPharma SPRL, Braine-l'Alleud, Belgium
| | - Erica A DePasquale
- Department of Biomedical Informatics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Khaled Alganem
- Department of Neuroscience, University of Toledo, Toledo, OH, USA
| | - Zhexing Wen
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Vahram Haroutunian
- Department of Psychiatry and Neuroscience, The Icahn School of Medicine at Mount Sinai, Bronx, NY, USA
| | - Pavel Katsel
- Department of Psychiatry and Neuroscience, The Icahn School of Medicine at Mount Sinai, Bronx, NY, USA
| | - Amy J Ramsey
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Jarek Meller
- Department of Biomedical Informatics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Electrical Engineering and Computer Science, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Informatics, Nicolaus Copernicus University, Torun, Poland
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | |
Collapse
|
46
|
Port RG, Dipiero MA, Ku M, Liu S, Blaskey L, Kuschner ES, Edgar JC, Roberts TP, Berman JI. Children with Autism Spectrum Disorder Demonstrate Regionally Specific Altered Resting-State Phase-Amplitude Coupling. Brain Connect 2019; 9:425-436. [PMID: 30900464 PMCID: PMC6588114 DOI: 10.1089/brain.2018.0653] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Studies suggest that individuals with autism spectrum disorder (ASD) exhibit altered electrophysiological alpha to gamma phase-amplitude coupling (PAC). Preliminary reports with small samples report conflicting findings regarding the directionality of the alpha to gamma PAC alterations in ASD. The present study examined resting-state activity throughout the brain in a relatively large sample of 119 children with ASD and 47 typically developing children. Children with ASD demonstrated regionally specific abnormalities in alpha to low-gamma PAC, with increased alpha to low-gamma PAC for a central midline source and decreased PAC at lateral sources. Group differences in local gamma-band power did not account for the regional group differences in alpha to low-gamma PAC. Moreover, local alpha power did not significantly modulate alpha to low-gamma PAC estimates. Finally, PAC estimates were correlated with Social Responsiveness Scale (SRS) indicating clinical relevance of the PAC metric. In conclusion, alpha to low-gamma PAC alterations in ASD demonstrate a heterogeneous spatial profile consistent with previous studies and were related to symptom severity.
Collapse
Affiliation(s)
- Russell G. Port
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Radiology, Lurie Family Foundations MEG Imaging Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Marissa A. Dipiero
- Department of Radiology, Lurie Family Foundations MEG Imaging Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Matthew Ku
- Department of Radiology, Lurie Family Foundations MEG Imaging Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Song Liu
- Department of Radiology, Lurie Family Foundations MEG Imaging Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Lisa Blaskey
- Department of Radiology, Lurie Family Foundations MEG Imaging Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, Center for Autism Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Emily S. Kuschner
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Radiology, Lurie Family Foundations MEG Imaging Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - J. Christopher Edgar
- Department of Radiology, Lurie Family Foundations MEG Imaging Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Timothy P.L. Roberts
- Department of Radiology, Lurie Family Foundations MEG Imaging Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jeffrey I. Berman
- Department of Radiology, Lurie Family Foundations MEG Imaging Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
47
|
Nakazawa H, Suzuki Y, Ishikawa Y, Bando Y, Yoshida S, Shiosaka S. Impaired social discrimination behavior despite normal social approach by kallikrein-related peptidase 8 knockout mouse. Neurobiol Learn Mem 2019; 162:47-58. [PMID: 31103466 DOI: 10.1016/j.nlm.2019.04.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 04/02/2019] [Accepted: 04/28/2019] [Indexed: 12/28/2022]
Abstract
For social mammals, recognition of conspecifics and discrimination of each other (social memory) is crucial to living in a stable colony. Here, we investigated whether kallikrein-related peptidase 8 (KLK8)-neuregulin 1 (NRG1)-ErbB signaling is crucial for social discrimination behavior using the social discrimination three chamber behavioral test. Klk8 knockout mice (NRG1-deactivated mice) exhibited normal social approach but impaired social discrimination. Intraventricular injection of recombinant NRG1177-246 into Klk8 knockout mice reversed this impaired social discrimination. This study reveals that KLK8 is a key regulator of NRG1-ErbB signaling, which contributes to social discrimination behavior.
Collapse
Affiliation(s)
- Hitomi Nakazawa
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Asahikawa, Hokkaido 078-8510, Japan.
| | - Yuka Suzuki
- Department of Systems Life Engineering, Maebashi Institute of Technology, Maebashi, Gunma 371-0816, Japan
| | - Yasuyuki Ishikawa
- Department of Systems Life Engineering, Maebashi Institute of Technology, Maebashi, Gunma 371-0816, Japan
| | - Yoshio Bando
- Department of Anatomy, Akita University Graduate School of Medicine, Akita, Akita 010-8543, Japan
| | - Shigetaka Yoshida
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Asahikawa, Hokkaido 078-8510, Japan
| | - Sadao Shiosaka
- Graduate School of Biological Science, Nara Institute of Science and Technology (NAIST), Ikoma, Nara 630-0192, Japan.
| |
Collapse
|
48
|
Port RG, Berman JI, Liu S, Featherstone RE, Roberts TP, Siegel SJ. Parvalbumin Cell Ablation of NMDA-R1 Leads to Altered Phase, But Not Amplitude, of Gamma-Band Cross-Frequency Coupling. Brain Connect 2019; 9:263-272. [PMID: 30588822 PMCID: PMC6479236 DOI: 10.1089/brain.2018.0639] [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] [Indexed: 12/22/2022] Open
Abstract
Altered gamma-band electrophysiological activity in individuals with autism spectrum disorder (ASD) is well documented, and analogous gamma-band alterations are recapitulated in several preclinical murine models relevant to ASD. Such gamma-band activity is hypothesized to underlie local circuit processes. Gamma-band cross-frequency coupling (CFC), a related though distinct metric, interrogates local neural circuit signal integration. Several recent studies have observed perturbed gamma-band CFC in individuals with ASD, although the direction of change remains unresolved. It also remains unclear whether murine models relevant to ASD recapitulate this altered gamma-band CFC. As such, this study examined whether mice with parvalbumin (PV) cell-specific ablation of NMDA-R1 (PVcre/NR1fl/fl) demonstrated altered gamma-band CFC as compared with their control littermates (PVcre/NR1+/+-mice that do not have the PV cell-specific ablation of NMDA-R1). Ten mice of each genotype had 4 min of "resting" electroencephalography recorded and analyzed. First, resting electrophysiological power was parsed into the canonical frequency bands and genotype-related differences were subsequently explored so as to provide context for the subsequent CFC analyses. PVcre/NR1fl/fl mice exhibited an increase in resting power specific to the high gamma-band, but not other frequency bands, as compared with PVcre/NR1+/+. CFC analyses then examined both the standard magnitude (strength) of CFC and the novel metric PhaseMax-which denotes the phase of the lower frequency signal at which the peak higher frequency signal power occurred. PVcre/NR1fl/fl mice exhibited altered PhaseMax, but not strength, of gamma-band CFC as compared with PVcre/NR1+/+ mice. As such, this study suggests a potential novel metric to explore when studying neuropsychiatric disorders.
Collapse
Affiliation(s)
- Russell G. Port
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Lurie Family Foundations MEG Imaging Center, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Jeffrey I. Berman
- Lurie Family Foundations MEG Imaging Center, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Song Liu
- Lurie Family Foundations MEG Imaging Center, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Robert E. Featherstone
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Timothy P.L. Roberts
- Lurie Family Foundations MEG Imaging Center, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Steven J. Siegel
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
49
|
Chronic unpredictable stress promotes cell-specific plasticity in prefrontal cortex D1 and D2 pyramidal neurons. Neurobiol Stress 2019; 10:100152. [PMID: 30937357 PMCID: PMC6430618 DOI: 10.1016/j.ynstr.2019.100152] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 03/04/2019] [Accepted: 03/04/2019] [Indexed: 11/24/2022] Open
Abstract
Exposure to unpredictable environmental stress is widely recognized as a major determinant for risk and severity in neuropsychiatric disorders such as major depressive disorder, anxiety, schizophrenia, and PTSD. The ability of ostensibly unrelated disorders to give rise to seemingly similar psychiatric phenotypes highlights a need to identify circuit-level concepts that could unify diverse factors under a common pathophysiology. Although difficult to disentangle a causative effect of stress from other factors on medial prefrontal cortex (PFC) dysfunction, a wealth of data from humans and rodents demonstrates that the PFC is a key target of stress. The present study sought to identify a model of chronic unpredictable stress (CUS) which induces affective behaviors in C57BL6J mice and once established, measure stress-related alterations in intrinsic excitability and synaptic regulation of mPFC layer 5/6 pyramidal neurons. Adult male mice received 2 weeks of 'less intense' stress or 2 or 4 weeks of 'more intense' CUS followed by sucrose preference for assessment of anhedonia, elevated plus maze for assessment of anxiety and forced swim test for assessment of depressive-like behaviors. Our findings indicate that more intense CUS exposure results in increased anhedonia, anxiety, and depressive behaviors, while the less intense stress results in no measured behavioral phenotypes. Once a behavioral model was established, mice were euthanized approximately 21 days post-stress for whole-cell patch clamp recordings from layer 5/6 pyramidal neurons in the prelimbic (PrL) and infralimbic (IL) cortices. No significant differences were initially observed in intrinsic cell excitability in either region. However, post-hoc analysis and subsequent confirmation using transgenic mice expressing tdtomato or eGFP under control of dopamine D1-or D2-type receptor showed that D1-expressing pyramidal neurons (D1-PYR) in the PrL exhibit reduced thresholds to fire an action potential (increased excitability) but impaired firing capacity at more depolarized potentials, whereas D2-expressing pyramidal neurons (D2-PYR) showed an overall reduction in excitability and spike firing frequency. Examination of synaptic transmission showed that D1-and D2-PYR exhibit differences in basal excitatory and inhibitory signaling under naïve conditions. In CUS mice, D1-PYR showed increased frequency of both miniature excitatory and inhibitory postsynaptic currents, whereas D2-PYR only showed a reduction in excitatory currents. These findings demonstrate that D1-and D2-PYR subpopulations differentially undergo stress-induced intrinsic and synaptic plasticity that may have functional implications for stress-related pathology, and that these adaptations may reflect unique differences in basal properties regulating output of these cells.
Collapse
|
50
|
Clinical trials in autism spectrum disorder: evidence, challenges and future directions. Curr Opin Neurol 2019; 31:119-125. [PMID: 29389748 DOI: 10.1097/wco.0000000000000542] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW The purpose of this manuscript is to review the evidence generated by clinical trials of pharmaceuticals in autism spectrum disorder (ASD), describe challenges in the conduct of such trials, and discuss future directions RECENT FINDINGS: Clinical trials in ASD have produced several compounds to adequately support the pharmacological treatment of associated symptom domains: attention deficit hyperactivity disorder (methylphenidate, atomoxetine, and alpha agonists), irritability/aggression (risperidone and aripiprazole), sleep (melatonin), and weight gain associated with atypical antipsychotic use (metformin). However, there is no evidence yet to support the routine use of pharmaceuticals for the treatment of core symptom domains. Challenges in the field include biological heterogeneity within ASD, lack of biomarkers that clarify biological heterogeneity or predict response to treatment, lack of data across the lifespan, and suboptimal outcome measures. SUMMARY Several compounds have evidence for the treatment of co-occurring symptoms in children and youth with ASD, although pharmacological interventions for core symptoms are still lacking. Identifying the various biologies underling ASD and developing biomarkers that stratify biologically homogeneous populations are both necessary to realize the promise of precision medicine in ASD.
Collapse
|