101
|
Alcaide J, Guirado R, Crespo C, Blasco-Ibáñez JM, Varea E, Sanjuan J, Nacher J. Alterations of perineuronal nets in the dorsolateral prefrontal cortex of neuropsychiatric patients. Int J Bipolar Disord 2019; 7:24. [PMID: 31728775 DOI: 10.1186/s40345-019-0161-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 10/08/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Alterations in the structure and physiology of interneurons in the prefrontal cortex (PFC) are important factors in the etiopathology of different psychiatric disorders. Among the interneuronal subpopulations, parvalbumin (PV) expressing cells appear to be specially affected. Interestingly, during development and adulthood the connectivity of these interneurons is regulated by the presence of perineuronal nets (PNNs), specialized regions of the extracellular matrix, which are frequently surrounding PV expressing neurons. Previous reports have found anomalies in the density of PNNs in the PFC of schizophrenic patients. However, although some studies have described alterations in PNNs in some extracortical regions of bipolar disorder patients, there are no studies focusing on the prefrontocortical PNNs of bipolar or major depression patients. For this reason, we have analyzed the density of PNNs in post-mortem sections of the dorsolateral PFC (DLPFC) from the Stanley Neuropathology Consortium, which includes controls, schizophrenia, bipolar and major depression patients. RESULTS We have not observed differences in the distribution of PV+ cells or PNNs, or in the percentage of PV+ interneurons surrounded by PNNs. The density of PV+ interneurons was similar in all the experimental groups, but there was a significantly lower density of PNNs in the DLPFC of bipolar disorder patients and a tendency towards a decrease in schizophrenic patients. No differences were found when evaluating the density of PV+ cells surrounded by PNNs. Interestingly, when assessing the influence of demographic data, we found an inverse correlation between the density of PNNs and the presence of psychosis. CONCLUSIONS The present results point to prefrontocortical PNNs and their role in the regulation of neuronal plasticity as putative players in the etiopathology of bipolar disorder and schizophrenia. Our findings also suggest a link between these specialized regions of the extracellular matrix and the presence of psychosis.
Collapse
Affiliation(s)
- Julia Alcaide
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de Valencia, Dr. Moliner 50, 46100, Burjassot, Spain
| | - Ramón Guirado
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de Valencia, Dr. Moliner 50, 46100, Burjassot, Spain
| | - Carlos Crespo
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de Valencia, Dr. Moliner 50, 46100, Burjassot, Spain
| | - José Miguel Blasco-Ibáñez
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de Valencia, Dr. Moliner 50, 46100, Burjassot, Spain
| | - Emilio Varea
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de Valencia, Dr. Moliner 50, 46100, Burjassot, Spain
| | - Julio Sanjuan
- Department of Medicine, Universitat de València, Valencia, Spain.,CIBERSAM: Spanish National Network for Research in Mental Health, Madrid, Spain.,Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| | - Juan Nacher
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de Valencia, Dr. Moliner 50, 46100, Burjassot, Spain. .,CIBERSAM: Spanish National Network for Research in Mental Health, Madrid, Spain. .,Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain.
| |
Collapse
|
102
|
Further delineation of neuropsychiatric findings in Tatton-Brown-Rahman syndrome due to disease-causing variants in DNMT3A: seven new patients. Eur J Hum Genet 2019; 28:469-479. [PMID: 31685998 DOI: 10.1038/s41431-019-0485-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/20/2019] [Accepted: 07/02/2019] [Indexed: 12/14/2022] Open
Abstract
Tatton-Brown-Rahman (TBRS) syndrome is a recently described overgrowth syndrome caused by loss of function variants in the DNMT3A gene. This gene encodes for a DNA methyltransferase 3 alpha, which is involved in epigenetic regulation, especially during embryonic development. Somatic variants in DNMT3A have been widely studied in different types of tumors, including acute myeloid leukemia, hematopoietic, and lymphoid cancers. Germline gain-of-function variants in this gene have been recently implicated in microcephalic dwarfism. Common clinical features of patients with TBRS include tall stature, macrocephaly, intellectual disability (ID), and a distinctive facial appearance. Differential diagnosis of TBRS comprises Sotos, Weaver, and Malan Syndromes. The majority of these disorders present other clinical features with a high clinical overlap, making necessary a molecular confirmation of the clinical diagnosis. We here describe seven new patients with variants in DNMT3A, four of them with neuropsychiatric disorders, including schizophrenia and psychotic behavior. In addition, one of the patients has developed a brain tumor in adulthood. This patient has also cerebral atrophy, aggressive behavior, ID, and abnormal facial features. Clinical evaluation of this group of patients should include a complete neuropsychiatric assessment together with psychological support in order to detect and manage abnormal behaviors such as aggressiveness, impulsivity, and attention deficit-hyperactivity disorder. TBRS should be suspected in patients with overgrowth, ID, tall stature, and macrocephaly, who also have some neuropsychiatric disorders without any genetic defects in the commonest overgrowth disorders. Molecular confirmation in these patients is mandatory.
Collapse
|
103
|
Trantham-Davidson H, Lavin A. Loss of dysbindin-1 affects GABAergic transmission in the PFC. Psychopharmacology (Berl) 2019; 236:3291-3300. [PMID: 31201475 PMCID: PMC6832803 DOI: 10.1007/s00213-019-05285-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 05/24/2019] [Indexed: 10/26/2022]
Abstract
It has been shown that dystrobrevin-binding protein 1 gene that encodes the protein dysbindin-1 is associated with risk for cognitive deficits, and studies have shown decreases in glutamate and correlated decreases in dysbindin-1 protein in the prefrontal cortex (PFC) and hippocampus of post-mortem tissue from schizophrenia patients. The PFC and the hippocampus have been shown to play a fundamental role in cognition, and studies in dysbindin-1 null mice have shown alterations in NMDAR located in pyramidal neurons as well as perturbation in LTP and cognitive deficits. The balance between excitatory and inhibitory transmission is crucial for normal cognitive functions; however, there is a dearth of information regarding the effects of loss of dysbindin-1 in GABAergic transmission. Using in vitro whole-cell clamp recordings, Western blots, and immunohistochemistry, we report here that dysbindin-1-deficient mice exhibit a significant decrease in the frequency of sIPSCs and in the amplitude of mIPSCs and significant decreases in PV staining and protein level. These results suggest that loss of dysbindin-1 affects GABAergic transmission at pre- and postsynaptic level and decreases parvalbumin markers.
Collapse
Affiliation(s)
| | - A Lavin
- Department of Neuroscience, MUSC, Charleston, SC, 29425, USA.
| |
Collapse
|
104
|
Dienel SJ, Lewis DA. Alterations in cortical interneurons and cognitive function in schizophrenia. Neurobiol Dis 2019; 131:104208. [PMID: 29936230 PMCID: PMC6309598 DOI: 10.1016/j.nbd.2018.06.020] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 05/31/2018] [Accepted: 06/20/2018] [Indexed: 12/18/2022] Open
Abstract
Certain clinical features of schizophrenia, such as working memory disturbances, appear to emerge from altered gamma oscillatory activity in the prefrontal cortex (PFC). Given the essential role of GABA neurotransmission in both working memory and gamma oscillations, understanding the cellular substrate for their disturbances in schizophrenia requires evidence from in vivo neuroimaging studies, which provide a means to link markers of GABA neurotransmission to gamma oscillations and working memory, and from postmortem studies, which provide insight into GABA neurotransmission at molecular and cellular levels of resolution. Here, we review findings from both types of studies which converge on the notions that 1) inhibitory GABA signaling in the PFC, especially between parvalbumin positive GABAergic basket cells and excitatory pyramidal cells, is required for gamma oscillatory activity and working memory function; and 2) disturbances in this signaling contribute to altered gamma oscillations and working memory in schizophrenia. Because the PFC is only one node in a distributed cortical network that mediates working memory, we also review evidence of GABA abnormalities in other cortical regions in schizophrenia.
Collapse
Affiliation(s)
- Samuel J Dienel
- Medical Scientist Training Program, University of Pittsburgh, United States; Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, United States
| | - David A Lewis
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, United States; Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, United States.
| |
Collapse
|
105
|
Imbriglio T, Verhaeghe R, Martinello K, Pascarelli MT, Chece G, Bucci D, Notartomaso S, Quattromani M, Mascio G, Scalabrì F, Simeone A, Maccari S, Del Percio C, Wieloch T, Fucile S, Babiloni C, Battaglia G, Limatola C, Nicoletti F, Cannella M. Developmental abnormalities in cortical GABAergic system in mice lacking mGlu3 metabotropic glutamate receptors. FASEB J 2019; 33:14204-14220. [PMID: 31665922 DOI: 10.1096/fj.201901093rrr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Polymorphic variants of the gene encoding for metabotropic glutamate receptor 3 (mGlu3) are linked to schizophrenia. Because abnormalities of cortical GABAergic interneurons lie at the core of the pathophysiology of schizophrenia, we examined whether mGlu3 receptors influence the developmental trajectory of cortical GABAergic transmission in the postnatal life. mGlu3-/- mice showed robust changes in the expression of interneuron-related genes in the prefrontal cortex (PFC), including large reductions in the expression of parvalbumin (PV) and the GluN1 subunit of NMDA receptors. The number of cortical cells enwrapped by perineuronal nets was increased in mGlu3-/- mice, suggesting that mGlu3 receptors shape the temporal window of plasticity of PV+ interneurons. Electrophysiological measurements of GABAA receptor-mediated responses revealed a more depolarized reversal potential of GABA currents in the somata of PFC pyramidal neurons in mGlu3-/- mice at postnatal d 9 associated with a reduced expression of the K+/Cl- symporter. Finally, adult mGlu3-/- mice showed lower power in electroencephalographic rhythms at 1-45 Hz in quiet wakefulness as compared with their wild-type counterparts. These findings suggest that mGlu3 receptors have a strong impact on the development of cortical GABAergic transmission and cortical neural synchronization mechanisms corroborating the concept that genetic variants of mGlu3 receptors may predispose to psychiatric disorders.-Imbriglio, T., Verhaeghe, R., Martinello, K., Pascarelli, M. T., Chece, G., Bucci, D., Notartomaso, S., Quattromani, M., Mascio, G., Scalabrì, F., Simeone, A., Maccari, S., Del Percio, C., Wieloch, T., Fucile, S., Babiloni, C., Battaglia, G., Limatola, C., Nicoletti, F., Cannella, M. Developmental abnormalities in cortical GABAergic system in mice lacking mGlu3 metabotropic glutamate receptors.
Collapse
Affiliation(s)
- Tiziana Imbriglio
- Department of Physiology and Pharmacology "V. Erspamer" University Sapienza of Rome, Rome, Italy
| | - Remy Verhaeghe
- Department of Physiology and Pharmacology "V. Erspamer" University Sapienza of Rome, Rome, Italy
| | - Katiuscia Martinello
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
| | - Maria Teresa Pascarelli
- Department of Physiology and Pharmacology "V. Erspamer" University Sapienza of Rome, Rome, Italy.,Oasi Research Institute - Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Troina, Italy
| | - Giuseppina Chece
- Department of Physiology and Pharmacology "V. Erspamer" University Sapienza of Rome, Rome, Italy
| | - Domenico Bucci
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
| | - Serena Notartomaso
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
| | - Miriana Quattromani
- Laboratory for Experimental Brain Research, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Giada Mascio
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
| | - Francesco Scalabrì
- Istituto di Ricerca Biologia Molecolare (IRBM) Science Park S.p.A., Pomezia, Rome, Italy
| | - Antonio Simeone
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", Centro Nazionale Ricerche (CNR), Naples, Italy
| | - Stefania Maccari
- Department of Science and Medical-Surgical Biotechnology, University Sapienza of Rome, Rome, Italy.,University of Lille, Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Claudio Del Percio
- Department of Physiology and Pharmacology "V. Erspamer" University Sapienza of Rome, Rome, Italy
| | - Tadeusz Wieloch
- Oasi Research Institute - Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Troina, Italy
| | - Sergio Fucile
- Department of Physiology and Pharmacology "V. Erspamer" University Sapienza of Rome, Rome, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
| | - Claudio Babiloni
- Department of Physiology and Pharmacology "V. Erspamer" University Sapienza of Rome, Rome, Italy.,Hospital San Raffaele Cassino, Cassino, Italy
| | - Giuseppe Battaglia
- Department of Physiology and Pharmacology "V. Erspamer" University Sapienza of Rome, Rome, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
| | - Cristina Limatola
- Department of Physiology and Pharmacology "V. Erspamer" University Sapienza of Rome, Rome, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology "V. Erspamer" University Sapienza of Rome, Rome, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
| | - Milena Cannella
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
| |
Collapse
|
106
|
Vostrikov VM. [Neuromorphological aspect of the GABAergic hypothesis of the pathogenesis of schizophrenia]. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 119:124-129. [PMID: 31626180 DOI: 10.17116/jnevro2019119081124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Many hypotheses have been proposed for the pathogenesis of schizophrenia. The most common hypotheses of schizophrenia are dopaminergic, serotoninergic, glutamatergic. There are also assumptions about involvement of other neurochemical systems, in particular GABAergic, in the pathogenesis of schizophrenia. The available data on the damage of GABAergic interneurons, taking into account the results of postmortem, neuroimaging, molecule-genetic, electrophysiological studies in humans and fundamental studies in animals, are discussed. The author suggests that one of the pathophysiological mechanisms of the pathogenesis of schizophrenia may be a disturbance of myelination of GABAergic interneurons leading to a decrease in the number of intra- and interhemispheric coherent connections, and eventually to the development of symptoms of the disease.
Collapse
|
107
|
Taylor SF, Grove TB, Ellingrod VL, Tso IF. The Fragile Brain: Stress Vulnerability, Negative Affect and GABAergic Neurocircuits in Psychosis. Schizophr Bull 2019; 45:1170-1183. [PMID: 31150555 PMCID: PMC6811817 DOI: 10.1093/schbul/sbz046] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Persons with schizophrenia exhibit sensitivity to stress and negative affect (NA), both strongly correlated with poor functional outcome. This theoretical review suggests that NA reflects a "fragile brain," ie, vulnerable to stress, including events not experienced as stressful by healthy individuals. Based on postmortem evidence of altered gamma-aminobutyric acid (GABA) function in parvalbumin positive interneurons (PVI), animal models of PVI abnormalities and neuroimaging data with GABAergic challenge, it is suggested that GABAergic disruptions weaken cortical regions, which leads to stress vulnerability and excessive NA. Neurocircuits that respond to stressful and salient environmental stimuli, such as the hypothalamic-pituitary-adrenal axis and the amygdala, are highly dysregulated in schizophrenia, exhibiting hypo- and hyper-activity. PVI abnormalities in lateral prefrontal cortex and hippocampus have been hypothesized to affect cognitive function and positive symptoms, respectively; in the medial frontal cortex (dorsal anterior cingulate cortex and dorsal medial prefrontal cortex), these abnormalities may lead to vulnerability to stress, NA and dysregulation of stress responsive systems. Given that postmortem PVI disruptions have been identified in other conditions, such as bipolar disorder and autism, stress vulnerability may reflect a transdiagnostic dimension of psychopathology.
Collapse
Affiliation(s)
- Stephan F Taylor
- Department of Psychiatry, University of Michigan, Rachel Upjohn Building, Ann Arbor, MI,To whom correspondence should be addressed; tel: 734-936-4955, fax: 734-936-7868, e-mail:
| | - Tyler B Grove
- Department of Psychiatry, University of Michigan, Rachel Upjohn Building, Ann Arbor, MI
| | | | - Ivy F Tso
- Department of Psychiatry, University of Michigan, Rachel Upjohn Building, Ann Arbor, MI
| |
Collapse
|
108
|
Agetsuma M, Hamm JP, Tao K, Fujisawa S, Yuste R. Parvalbumin-Positive Interneurons Regulate Neuronal Ensembles in Visual Cortex. Cereb Cortex 2019; 28:1831-1845. [PMID: 29106504 DOI: 10.1093/cercor/bhx169] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 06/16/2017] [Indexed: 01/20/2023] Open
Abstract
For efficient cortical processing, neural circuit dynamics must be spatially and temporally regulated with great precision. Although parvalbumin-positive (PV) interneurons can control network synchrony, it remains unclear how they contribute to spatio-temporal patterning of activity. We investigated this by optogenetic inactivation of PV cells with simultaneous two-photon Ca2+ imaging from populations of neurons in mouse visual cortex in vivo. For both spontaneous and visually evoked activity, PV interneuron inactivation decreased network synchrony. But, interestingly, the response reliability and spatial extent of coactive neuronal ensembles during visual stimulation were also disrupted by PV-cell suppression, which reduced the functional repertoire of ensembles. Thus, PV interneurons can control the spatio-temporal dynamics of multineuronal activity by functionally sculpting neuronal ensembles and making them more different from each other. In doing so, inhibitory circuits could help to orthogonalize multicellular patterns of activity, enabling neural circuits to more efficiently occupy a higher dimensional space of potential dynamics.
Collapse
Affiliation(s)
- Masakazu Agetsuma
- Neurotechnology Center, Department of Biological Sciences, Columbia University, 550 West 120 Street, Box 4822, New York, NY 10027, USA.,Japan Science and Technology Agency, PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan.,The Institute of Scientific and Industrial Research, Osaka University, Mihogaoka 8-1, Ibaraki, Osaka 567-0047, Japan.,National Institute for Physiological Sciences, Division of Homeostatic Development, 38 Nishigohnaka Myodaiji-cho, Okazaki, Aichi 444-8585, Japan
| | - Jordan P Hamm
- Neurotechnology Center, Department of Biological Sciences, Columbia University, 550 West 120 Street, Box 4822, New York, NY 10027, USA
| | - Kentaro Tao
- RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama351-0106, Japan
| | | | - Rafael Yuste
- Neurotechnology Center, Department of Biological Sciences, Columbia University, 550 West 120 Street, Box 4822, New York, NY 10027, USA
| |
Collapse
|
109
|
Seney ML, Cahill K, Enwright JF, Logan RW, Huo Z, Zong W, Tseng G, McClung CA. Diurnal rhythms in gene expression in the prefrontal cortex in schizophrenia. Nat Commun 2019; 10:3355. [PMID: 31399567 PMCID: PMC6689017 DOI: 10.1038/s41467-019-11335-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 06/25/2019] [Indexed: 01/06/2023] Open
Abstract
Schizophrenia is associated with disrupted cognitive control and sleep-wake cycles. Here we identify diurnal rhythms in gene expression in the human dorsolateral prefrontal cortex (dlPFC), in schizophrenia and control subjects. We find significant diurnal (24 h) rhythms in control subjects, however, most of these transcripts are not rhythmic in subjects with schizophrenia. Instead, subjects with schizophrenia have a different set of rhythmic transcripts. The top pathways identified in transcripts rhythmic only in subjects with schizophrenia are associated with mitochondrial function. Importantly, these rhythms drive differential expression patterns of these and several other genes that have long been implicated in schizophrenia (including BDNF and GABAergic-related transcripts). Indeed, differential expression of these transcripts is only seen in subjects that died during the night, with no change in subjects that died during the day. These data provide insights into a potential mechanism that underlies changes in gene expression in the dlPFC with schizophrenia.
Collapse
Affiliation(s)
- Marianne L Seney
- Translational Neuroscience Program, Department of Psychiatry, Center for Neuroscience, University of Pittsburgh, Pittsburgh, 15213, PA, USA
| | - Kelly Cahill
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, 15261, PA, USA
| | - John F Enwright
- Translational Neuroscience Program, Department of Psychiatry, Center for Neuroscience, University of Pittsburgh, Pittsburgh, 15213, PA, USA
| | - Ryan W Logan
- Translational Neuroscience Program, Department of Psychiatry, Center for Neuroscience, University of Pittsburgh, Pittsburgh, 15213, PA, USA
| | - Zhiguang Huo
- Department of Biostatistics, University of Florida, Gainesville, 32611, FL, USA
| | - Wei Zong
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, 15261, PA, USA
| | - George Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, 15261, PA, USA
| | - Colleen A McClung
- Translational Neuroscience Program, Department of Psychiatry, Center for Neuroscience, University of Pittsburgh, Pittsburgh, 15213, PA, USA.
| |
Collapse
|
110
|
Duchatel RJ, Shannon Weickert C, Tooney PA. White matter neuron biology and neuropathology in schizophrenia. NPJ SCHIZOPHRENIA 2019; 5:10. [PMID: 31285426 PMCID: PMC6614474 DOI: 10.1038/s41537-019-0078-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/06/2019] [Indexed: 12/17/2022]
Abstract
Schizophrenia is considered a neurodevelopmental disorder as it often manifests before full brain maturation and is also a cerebral cortical disorder where deficits in GABAergic interneurons are prominent. Whilst most neurons are located in cortical and subcortical grey matter regions, a smaller population of neurons reside in white matter tracts of the primate and to a lesser extent, the rodent brain, subjacent to the cortex. These interstitial white matter neurons (IWMNs) have been identified with general markers for neurons [e.g., neuronal nuclear antigen (NeuN)] and with specific markers for neuronal subtypes such as GABAergic neurons. Studies of IWMNs in schizophrenia have primarily focused on their density underneath cortical areas known to be affected in schizophrenia such as the dorsolateral prefrontal cortex. Most of these studies of postmortem brains have identified increased NeuN+ and GABAergic IWMN density in people with schizophrenia compared to healthy controls. Whether IWMNs are involved in the pathogenesis of schizophrenia or if they are increased because of the cortical pathology in schizophrenia is unknown. We also do not understand how increased IWMN might contribute to brain dysfunction in the disorder. Here we review the literature on IWMN pathology in schizophrenia. We provide insight into the postulated functional significance of these neurons including how they may contribute to the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Ryan J Duchatel
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia
- Priority Centre for Brain and Mental Health Research and Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Cynthia Shannon Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, 2031, Australia
- School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, 2052, Australia
- Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, New York, 13210, USA
| | - Paul A Tooney
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia.
- Priority Centre for Brain and Mental Health Research and Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, 2308, Australia.
| |
Collapse
|
111
|
Postmortem transcriptional profiling reveals widespread increase in inflammation in schizophrenia: a comparison of prefrontal cortex, striatum, and hippocampus among matched tetrads of controls with subjects diagnosed with schizophrenia, bipolar or major depressive disorder. Transl Psychiatry 2019; 9:151. [PMID: 31123247 PMCID: PMC6533277 DOI: 10.1038/s41398-019-0492-8] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 05/03/2019] [Indexed: 11/30/2022] Open
Abstract
Psychiatric disorders such as schizophrenia (SCZ), bipolar disorder (BD), and major depressive disorder (MDD) arise from complex interactions between genetic and environmental factors. Common genetic variants associated with multiple psychiatric disorders suggest that shared genetic architecture could contribute to divergent clinical syndromes. To evaluate shared transcriptional alterations across connected brain regions, Affymetrix microarrays were used to profile postmortem dorsolateral prefrontal cortex (DLPFC), hippocampus, and associative striatum from 19 well-matched tetrads of subjects with SCZ, BD, MDD, or unaffected controls. SCZ subjects showed a substantial burden of differentially expressed genes across all examined brain regions with the greatest effects in hippocampus, whereas BD and MDD showed less robust alterations. Pathway analysis of transcriptional profiles compared across diagnoses demonstrated commonly enriched pathways between all three disorders in hippocampus, significant overlap between SCZ and BD in DLPFC, but no significant overlap of enriched pathways between disorders in striatum. SCZ samples showed increased expression of transcripts associated with inflammation across all brain regions examined, which was not evident in BD or MDD, or in rat brain following chronic dosing with antipsychotic drugs. Several markers of inflammation were confirmed by RT-PCR in hippocampus, including S100A8/9, IL-6, MAFF, APOLD1, IFITM3, and BAG3. A cytokine ELISA panel showed significant increases in IL-2 and IL-12p70 protein content in hippocampal tissue collected from same SCZ subjects when compared to matched control subjects. These data suggest an overlapping subset of dysregulated pathways across psychiatric disorders; however, a widespread increase in inflammation appears to be a specific feature of the SCZ brain and is not likely to be attributable to chronic antipsychotic drug treatment.
Collapse
|
112
|
Short-Term Exposure to Enriched Environment in Adult Rats Restores MK-801-Induced Cognitive Deficits and GABAergic Interneuron Immunoreactivity Loss. Mol Neurobiol 2019; 55:26-41. [PMID: 28822057 DOI: 10.1007/s12035-017-0715-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Perinatal injections of N-methyl-D-aspartate (NMDA) receptor antagonist in rodents emulate some cognitive impairments and neurochemical alterations, such as decreased GABAergic (gamma aminobutyric acid) interneuron immunoreactivity, also found in schizophrenia. These features are pervasive, and developing neuroprotective or neurorestorative strategies is of special interest. In this work, we aimed to investigate if a short exposure to enriched environment (EE) in early adulthood (P55-P73) was an effective strategy to improve cognitive dysfunction and to restore interneuron expression in medial prefrontal cortex (mPFC) and hippocampus (HPC). For that purpose, we administered MK-801 intraperitoneally to Long Evans rats from postnatal days 10 to 20. Twenty-four hours after the last injection, MK-801 produced a transient decrease in spontaneous motor activity and exploration, but those abnormalities were absent at P24 and P55. The open field test on P73 manifested that EE reduced anxiety-like behavior. In addition, MK-801-treated rats showed cognitive impairment in novel object recognition test that was reversed by EE. We quantified different interneuron populations based on their calcium-binding protein expression (parvalbumin, calretinin, and calbindin), glutamic acid decarboxylase 67, and neuronal nuclei-positive cells by means of unbiased stereology and found that EE enhanced interneuron immunoreactivity up to normal values in MK-801-treated rats. Our results demonstrate that a timely intervention with EE is a powerful tool to reverse long-lasting changes in cognition and neurochemical markers of interneurons in an animal model of schizophrenia.
Collapse
|
113
|
Pathogenic potential of human SLC12A5 variants causing KCC2 dysfunction. Brain Res 2019; 1710:1-7. [DOI: 10.1016/j.brainres.2018.12.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/21/2018] [Accepted: 12/17/2018] [Indexed: 12/29/2022]
|
114
|
Trakadis YJ, Sardaar S, Chen A, Fulginiti V, Krishnan A. Machine learning in schizophrenia genomics, a case-control study using 5,090 exomes. Am J Med Genet B Neuropsychiatr Genet 2019; 180:103-112. [PMID: 29704323 DOI: 10.1002/ajmg.b.32638] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 02/28/2018] [Accepted: 03/30/2018] [Indexed: 12/21/2022]
Abstract
Our hypothesis is that machine learning (ML) analysis of whole exome sequencing (WES) data can be used to identify individuals at high risk for schizophrenia (SCZ). This study applies ML to WES data from 2,545 individuals with SCZ and 2,545 unaffected individuals, accessed via the database of genotypes and phenotypes (dbGaP). Single nucleotide variants and small insertions and deletions were annotated by ANNOVAR using the reference genome hg19/GRCh37. Rare (predicted functional) variants with a minor allele frequency ≤1% and genotype quality ≥90 including missense, frameshift, stop gain, stop loss, intronic, and exonic splicing variants were selected. A file containing all cases and controls, the names of genes with variants meeting our criteria, and the number of variants per gene for each individual, was used for ML analysis. The supervised machine-learning algorithm used the patterns of variants observed in the different genes to determine which subset of genes can best predict that an individual is affected. Seventy percent of the data was used to train the algorithm and the remaining 30% of data (n = 1,526) was used to evaluate its efficiency. The supervised ML algorithm, gradient boosted trees with regularization (eXtreme Gradient Boosting implementation) was the best performing algorithm yielding promising results (accuracy: 85.7%, specificity: 86.6%, sensitivity: 84.9%, area under the receiver-operator characteristic curve: 0.95). The top 50 features (genes) of the algorithm were analyzed using bioinformatics resources for new insights about the pathophysiology of SCZ. This manuscript presents a novel predictor which could potentially enable studies exploring disease-modifying intervention in the early stages of the disease.
Collapse
Affiliation(s)
- Yannis J Trakadis
- Department of Human Genetics, McGill University, Montreal, Québec, Canada
| | - Sameer Sardaar
- Department of Human Genetics, McGill University, Montreal, Québec, Canada
| | - Anthony Chen
- Department of Human Genetics, McGill University, Montreal, Québec, Canada
| | - Vanessa Fulginiti
- Department of Human Genetics, McGill University, Montreal, Québec, Canada
| | - Ankur Krishnan
- Department of Human Genetics, McGill University, Montreal, Québec, Canada
| |
Collapse
|
115
|
Selective Activation of Cholecystokinin-Expressing GABA (CCK-GABA) Neurons Enhances Memory and Cognition. eNeuro 2019; 6:eN-NWR-0360-18. [PMID: 30834305 PMCID: PMC6397954 DOI: 10.1523/eneuro.0360-18.2019] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/04/2019] [Accepted: 01/23/2019] [Indexed: 12/15/2022] Open
Abstract
Cholecystokinin-expressing GABAergic (CCK-GABA) neurons are perisomatic inhibitory cells that have been argued to regulate emotion and sculpt the network oscillations associated with cognition. However, no study has selectively manipulated CCK-GABA neuron activity during behavior in freely-moving animals. To explore the behavioral effects of activating CCK-GABA neurons on emotion and cognition, we utilized a novel intersectional genetic mouse model coupled with a chemogenetic approach. Specifically, we generated triple transgenic CCK-Cre;Dlx5/6-Flpe;RC::FL-hM3Dq (CCK-GABA/hM3Dq) mice that expressed the synthetic excitatory hM3Dq receptor in CCK-GABA neurons. Results showed that clozapine-N-oxide (CNO)-mediated activation of CCK-GABA neurons did not alter open field (OF) or tail suspension (TS) performance and only slightly increased anxiety in the elevated plus maze (EPM). Although CNO treatment had only modestly affected emotional behavior, it significantly enhanced multiple cognitive and memory behaviors including social recognition, contextual fear conditioning, contextual discrimination, object recognition, and problem-solving in the puzzle box. Collectively, these findings suggest that systemic activation of CCK-GABA neurons minimally affects emotion but significantly enhances cognition and memory. Our results imply that CCK-GABA neurons are more functionally diverse than originally expected and could serve as a potential therapeutic target for the treatment of cognitive/memory disorders.
Collapse
|
116
|
Impaired Interneuron Development in a Novel Model of Neonatal Brain Injury. eNeuro 2019; 6:eN-NWR-0300-18. [PMID: 30809588 PMCID: PMC6390196 DOI: 10.1523/eneuro.0300-18.2019] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 12/27/2018] [Accepted: 01/03/2019] [Indexed: 12/20/2022] Open
Abstract
Prematurity is associated with significantly increased risk of neurobehavioral pathologies, including autism and schizophrenia. A common feature of these psychiatric disorders is prefrontal cortex (PFC) inhibitory circuit disruption due to GABAergic interneuron alteration. Cortical interneurons are generated and migrate throughout late gestation and early infancy, making them highly susceptible to perinatal insults such as preterm birth. Term and preterm PFC pathology specimens were assessed using immunohistochemical markers for interneurons. Based on the changes seen, a new preterm encephalopathy mouse model was developed to produce similar PFC interneuron loss. Maternal immune activation (MIA; modeling chorioamnionitis, associated with 85% of extremely preterm births) was combined with chronic sublethal hypoxia (CSH; modeling preterm respiratory failure), with offspring of both sexes assessed anatomically, molecularly and neurobehaviorally. In the PFC examined from the human preterm samples compared to matched term samples at corrected age, a decrease in somatostatin (SST) and calbindin (CLB) interneurons was seen in upper cortical layers. This pattern of interneuron loss in upper cortical layers was mimicked in the mouse PFC following the combination of MIA and CSH, but not after either insult alone. This persistent interneuron loss is associated with postnatal microglial activation that occurs during CSH only after MIA. The combined insults lead to long-term neurobehavioral deficits which parallel human psychopathologies that may be seen after extremely preterm birth. This new preclinical model supports a paradigm in which specific cellular alterations seen in preterm encephalopathy can be linked with a risk of neuropsychiatric sequela. Specific interneuron subtypes may provide therapeutic targets to prevent or ameliorate these neurodevelopmental risks.
Collapse
|
117
|
Transcriptomic immaturity inducible by neural hyperexcitation is shared by multiple neuropsychiatric disorders. Commun Biol 2019; 2:32. [PMID: 30675529 PMCID: PMC6342824 DOI: 10.1038/s42003-018-0277-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 12/13/2018] [Indexed: 02/07/2023] Open
Abstract
Biomarkers are needed to improve the diagnosis of neuropsychiatric disorders, which are often associated to excitatory/inhibitory imbalances in neural transmission and abnormal maturation. Here, we characterized different disease conditions by mapping changes in the expression patterns of maturation-related genes whose expression was altered by experimental neural hyperexcitation in published studies. This analysis revealed two gene expression patterns: decreases in maturity markers and increases in immaturity markers. These two groups of genes were characterized by the over-representation of genes related to synaptic function and chromosomal modification, respectively. Using these two groups in a transdiagnostic analysis of 87 disease datasets for eight neuropsychiatric disorders and 12 datasets from corresponding animal models, we found that transcriptomic pseudoimmaturity inducible by neural hyperexcitation is shared by multiple neuropsychiatric disorders, such as schizophrenia, Alzheimer disorders, and amyotrophic lateral sclerosis. Our results indicate that this endophenotype serves as a basis for the transdiagnostic characterization of these disorders. Tomoyuki Murano et al. showed that neural hyperexcitation increases the expression of immaturity related genes. These changes in gene expression are shared among different neuropsychiatric and neurological conditions, hinting at their potential role as biomarkers.
Collapse
|
118
|
Pandya M, Palpagama TH, Turner C, Waldvogel HJ, Faull RL, Kwakowsky A. Sex- and age-related changes in GABA signaling components in the human cortex. Biol Sex Differ 2019; 10:5. [PMID: 30642393 PMCID: PMC6332906 DOI: 10.1186/s13293-018-0214-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 12/09/2018] [Indexed: 12/13/2022] Open
Abstract
Gamma-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the nervous system. Previous studies have shown fluctuations in expression levels of GABA signaling components-glutamic acid decarboxylase (GAD), GABA receptor (GABAR) subunit, and GABA transporter (GAT)-with increasing age and between sexes; however, this limited knowledge is highly based on animal models that produce inconsistent findings. This study is the first analysis of the age- and sex-specific changes of the GAD, GABAA/BR subunits, and GAT expression in the human primary sensory and motor cortices; superior (STG), middle (MTG), and inferior temporal gyrus (ITG); and cerebellum. Utilizing Western blotting, we found that the GABAergic system is relatively robust against sex and age-related differences in all brain regions examined. However, we observed several sex-dependent differences in GABAAR subunit expression in STG along with age-dependent GABAAR subunit and GAD level alteration. No significant age-related differences were found in α1, α2, α5, β3, and γ2 subunit expression in the STG. However, we found significantly higher GABAAR α3 subunit expression in the STG in young males compared to old males. We observed a significant sex-dependent difference in α1 subunit expression: males presenting significantly higher levels compared to women across all stages of life in STG. Older females showed significantly lower α2, α5, and β3 subunit expression compared to old males in the STG. These changes found in the STG might significantly influence GABAergic neurotransmission and lead to sex- and age-specific disease susceptibility and progression.
Collapse
Affiliation(s)
- Madhavi Pandya
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Thulani H. Palpagama
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Clinton Turner
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Anatomical Pathology, LabPlus, Auckland City Hospital, Auckland, New Zealand
| | - Henry J. Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard L. Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
119
|
Gomes FV, Edelson JR, Volk DW, Grace AA. Altered brain cannabinoid 1 receptor mRNA expression across postnatal development in the MAM model of schizophrenia. Schizophr Res 2018; 201:254-260. [PMID: 29705007 PMCID: PMC6203675 DOI: 10.1016/j.schres.2018.04.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 04/17/2018] [Accepted: 04/19/2018] [Indexed: 12/14/2022]
Abstract
Altered cannabinoid 1 receptor (CB1R) expression has been reported in the brain of subjects with schizophrenia, a developmental mental illness that usually emerges in late adolescence/early adulthood. However, the developmental period at which changes in the CB1R expression appear in schizophrenia is unknown. To gain insight into this factor, we assessed the postnatal developmental trajectory of CB1R expression in the methylazoxymethanol (MAM) model of schizophrenia. Using in situ hybridization with film and grain analyses, CB1R messenger RNA (mRNA) levels were quantified in multiple brain regions, including the medial prefrontal cortex (mPFC), secondary motor cortex, dorsomedial and dorsolateral striatum, dorsal subregions and ventral subiculum of the hippocampus, of MAM-treated rats and normal controls at three developmental periods [juvenile - postnatal day (PD) 30; adolescence - PD45; and adulthood - PD85]. In all brain regions studied, CB1R mRNA levels were highest in juveniles and then decreased progressively toward adolescent and adult levels in control and MAM-treated rats. However, in MAM-treated rats, CB1R mRNA levels were lower in the mPFC at PD85 and higher in the dorsolateral striatum at PD45 and PD85 relative to controls. Cellular analyses confirmed the changes in CB1R mRNA expression in MAM-treated rats. These findings are in accordance with previous studies showing a decrease in the CB1R mRNA expression from juvenile period to adolescence to adulthood in cortical, striatal, and hippocampal regions. Additionally, similar to most of the schizophrenia-like signs observed in the MAM model, embryonic exposure to MAM leads to schizophrenia-related changes in CB1R mRNA expression that only emerge later in development.
Collapse
Affiliation(s)
- Felipe V Gomes
- Department of Neuroscience, University of Pittsburgh, PA, USA.
| | | | - David W Volk
- Department of Psychiatry, University of Pittsburgh, PA, USA
| | - Anthony A Grace
- Department of Neuroscience, University of Pittsburgh, PA, USA; Department of Psychiatry, University of Pittsburgh, PA, USA; Department of Psychology, University of Pittsburgh, PA, USA
| |
Collapse
|
120
|
Abbas AI, Sundiang MJM, Henoch B, Morton MP, Bolkan SS, Park AJ, Harris AZ, Kellendonk C, Gordon JA. Somatostatin Interneurons Facilitate Hippocampal-Prefrontal Synchrony and Prefrontal Spatial Encoding. Neuron 2018; 100:926-939.e3. [PMID: 30318409 DOI: 10.1016/j.neuron.2018.09.029] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 07/30/2018] [Accepted: 09/18/2018] [Indexed: 01/04/2023]
Abstract
Decreased hippocampal-prefrontal synchrony may mediate cognitive deficits in schizophrenia, but it remains unclear which cells orchestrate this long-range synchrony. Parvalbumin (PV)- and somatostatin (SOM)-expressing interneurons show histological abnormalities in individuals with schizophrenia and are hypothesized to regulate oscillatory synchrony within the prefrontal cortex. To examine the relationship between interneuron function, long-range hippocampal-prefrontal synchrony, and cognition, we optogenetically inhibited SOM and PV neurons in the medial prefrontal cortex (mPFC) of mice performing a spatial working memory task while simultaneously recording neural activity in the mPFC and the hippocampus (HPC). We found that inhibiting SOM, but not PV, interneurons during the encoding phase of the task impaired working memory accuracy. This behavioral impairment was associated with decreased hippocampal-prefrontal synchrony and impaired spatial encoding in mPFC neurons. These findings suggest that interneuron dysfunction may contribute to cognitive deficits associated with schizophrenia by disrupting long-range synchrony between the HPC and PFC.
Collapse
Affiliation(s)
- Atheir I Abbas
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Marina J M Sundiang
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Britt Henoch
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Mitchell P Morton
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Scott S Bolkan
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Alan J Park
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Alexander Z Harris
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Christoph Kellendonk
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Joshua A Gordon
- National Institute of Mental Health, Bethesda, MD 20892, USA.
| |
Collapse
|
121
|
Kirenskaya AV, Storozheva ZI, Gruden MA, Sewell RDE. COMT and GAD1 gene polymorphisms are associated with impaired antisaccade task performance in schizophrenic patients. Eur Arch Psychiatry Clin Neurosci 2018; 268:571-584. [PMID: 29429137 PMCID: PMC6096577 DOI: 10.1007/s00406-018-0881-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 02/04/2018] [Indexed: 12/19/2022]
Abstract
Genetic influences modulating executive functions engaging prefrontal cortical brain systems were investigated in 141 male subjects. The effects of variations in two genes implicated in dopamine and GABA activities in the prefrontal cortex: rs4680 (Val158/Met polymorphism of the catechol-o-methyltransferase gene-COMT) and rs3749034 (C/T) substitution in the promoter region of the glutamic acid decarboxylase gene (GAD1) were studied on antisaccade (AS) performance in healthy subjects and schizophrenic patients. Genotyping revealed a trend towards a reduced proportion of COMT Val/Met heterozygotes and a significantly increased frequency of the GAD1 rs3749034 C allele in schizophrenic patients relative to controls. Patients had elevated error rates, increased AS latencies and increased latency variability (coefficient of variation) compared to controls. The influence of polymorphisms was observed only in patients but not in controls. A substantial effect of the COMT genotype was noted on the coefficient of variation in latency, and this measure was higher in Val homozygotes compared to Met allele carriers (p < 0.05) in the patient group. The outcome from rs3749034 was also disclosed on the error rate (higher in T carriers relative to C homozygotes, p < 0.01) and latency (increased in C homozygotes relative to T carriers, p < 0.01). Binary logistic regression showed that inclusion of the genotype factor (i.e., selective estimation of antisaccade measures in CC carriers) considerably increased the validity of the diagnostic model based on the AS measures. These findings may well be derived from specific genetic associations with prefrontal cortex functioning in schizophrenia.
Collapse
Affiliation(s)
- Anna V Kirenskaya
- Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky Lane. 23, 119034, Moscow, Russian Federation
| | - Zinaida I Storozheva
- Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky Lane. 23, 119034, Moscow, Russian Federation
| | - Marina A Gruden
- Federal State Budgetary Scientific Institution "P. K. Anokhin Research Institute of Normal Physiology", Baltiskaya St., 8, 125315, Moscow, Russian Federation
| | - Robert D E Sewell
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Redwood Building, Cardiff University, Cardiff, CF10 3NB, UK.
| |
Collapse
|
122
|
Andersson JD, Matuskey D, Finnema SJ. Positron emission tomography imaging of the γ-aminobutyric acid system. Neurosci Lett 2018; 691:35-43. [PMID: 30102960 DOI: 10.1016/j.neulet.2018.08.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 08/06/2018] [Accepted: 08/09/2018] [Indexed: 01/08/2023]
Abstract
In this review, we summarize the recent development of positron emission tomography (PET) radioligands for γ-aminobutyric acid A (GABAA) receptors and their potential to measure changes in endogenous GABA levels and highlight the clinical and translational applications of GABA-sensitive PET radioligands. We review the basic physiology of the GABA system with a focus on the importance of GABAA receptors in the brain and specifically the benzodiazepine binding site. Challenges for the development of central nervous system radioligands and particularly for radioligands with increased GABA sensitivity are outlined, as well as the status of established benzodiazepine site PET radioligands and agonist GABAA radioligands. We underline the challenge of using allosteric interactions to measure GABA concentrations and review the current state of PET imaging of changes in GABA levels. We conclude that PET tracers with increased GABA sensitivity are required to efficiently measure GABA release and that such a tool could be broadly applied to assess GABA transmission in vivo across several disorders.
Collapse
Affiliation(s)
- Jan D Andersson
- University of Alberta, Medical Isotope and Cyclotron Facility, Edmonton, Canada
| | - David Matuskey
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Sjoerd J Finnema
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA; Center for Psychiatric Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
123
|
Wen TH, Binder DK, Ethell IM, Razak KA. The Perineuronal 'Safety' Net? Perineuronal Net Abnormalities in Neurological Disorders. Front Mol Neurosci 2018; 11:270. [PMID: 30123106 PMCID: PMC6085424 DOI: 10.3389/fnmol.2018.00270] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 07/17/2018] [Indexed: 12/22/2022] Open
Abstract
Perineuronal nets (PNN) are extracellular matrix (ECM) assemblies that preferentially ensheath parvalbumin (PV) expressing interneurons. Converging evidence indicates that PV cells and PNN are impaired in a variety of neurological disorders. PNN development and maintenance is necessary for a number of processes within the CNS, including regulation of GABAergic cell function, protection of neurons from oxidative stress, and closure of developmental critical period plasticity windows. Understanding PNN functions may be essential for characterizing the mechanisms of altered cortical excitability observed in neurodegenerative and neurodevelopmental disorders. Indeed, PNN abnormalities have been observed in post-mortem brain tissues of patients with schizophrenia and Alzheimer’s disease. There is impaired development of PNNs and enhanced activity of its key regulator matrix metalloproteinase-9 (MMP-9) in Fragile X Syndrome, a common genetic cause of autism. MMP-9, a protease that cleaves ECM, is differentially regulated in a number of these disorders. Despite this, few studies have addressed the interactions between PNN expression, MMP-9 activity and neuronal excitability. In this review, we highlight the current evidence for PNN abnormalities in CNS disorders associated with altered network function and MMP-9 levels, emphasizing the need for future work targeting PNNs in pathophysiology and therapeutic treatment of neurological disorders.
Collapse
Affiliation(s)
- Teresa H Wen
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States
| | - Devin K Binder
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States.,Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Iryna M Ethell
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States.,Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Khaleel A Razak
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States.,Psychology Graduate Program, Department of Psychology, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
124
|
Prefrontal cortex-dependent innate behaviors are altered by selective knockdown of Gad1 in neuropeptide Y interneurons. PLoS One 2018; 13:e0200809. [PMID: 30024942 PMCID: PMC6053188 DOI: 10.1371/journal.pone.0200809] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 06/09/2018] [Indexed: 12/23/2022] Open
Abstract
GABAergic dysfunction has been implicated in a variety of neurological and psychiatric disorders, including anxiety disorders. Anxiety disorders are the most common type of psychiatric disorder during adolescence. There is a deficiency of GABAergic transmission in anxiety, and enhancement of GABA transmission through pharmacological means reduces anxiety behaviors. GAD67—the enzyme responsible for GABA production–has been linked to anxiety disorders. One class of GABAergic interneurons, Neuropeptide Y (NPY) expressing cells, is abundantly found in brain regions associated with anxiety and fear learning, including prefrontal cortex, hippocampus and amygdala. Additionally, NPY itself has been shown to have anxiolytic effects, and loss of NPY+ interneurons enhances anxiety behaviors. A previous study showed that knockdown of Gad1 from NPY+ cells led to reduced anxiety behaviors in adult mice. However, the role of GABA release from NPY+ interneurons in adolescent anxiety is unclear. Here we used a transgenic mouse that reduces GAD67 in NPY+ cells (NPYGAD1-TG) through Gad1 knockdown and tested for effects on behavior in adolescent mice. Adolescent NPYGAD1-TG mice showed enhanced anxiety-like behavior and sex-dependent changes in locomotor activity. We also found enhancement in two other innate behavioral tasks, nesting construction and social dominance. In contrast, fear learning was unchanged. Because we saw changes in behavioral tasks dependent upon prefrontal cortex and hippocampus, we investigated the extent of GAD67 knockdown in these regions. Immunohistochemistry revealed a 40% decrease in GAD67 in NPY+ cells in prefrontal cortex, indicating a significant but incomplete knockdown of GAD67. In contrast, there was no decrease in GAD67 in NPY+ cells in hippocampus. Consistent with this, there was no change in inhibitory synaptic transmission in hippocampus. Our results show the behavioral impact of cell-specific interneuron dysfunction and suggest that GABA release by NPY+ cells is important for regulating innate prefrontal cortex-dependent behavior in adolescents.
Collapse
|
125
|
Zhang Y, Catts VS, Shannon Weickert C. Lower antioxidant capacity in the prefrontal cortex of individuals with schizophrenia. Aust N Z J Psychiatry 2018; 52:690-698. [PMID: 28891319 DOI: 10.1177/0004867417728805] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVE The glutathione (GSH) pathway is the main antioxidant system to protect against oxidative stress in the human brain. In this study, we tested whether molecular components of the GSH antioxidant system are changed in dorsolateral prefrontal cortex tissue from people with schizophrenia compared to controls. METHOD The levels of total glutathione and reduced GSH were determined by fluorometric assay via quantifying thiols in extracts from frontal cortex of 68 people. Immunoblotting was used to measure levels of enzymes responsible for maintaining GSH, the glutamyl-cysteine ligase (GCL) catalytic subunit (GCLC) and the GSH peroxidase (GPx)-like protein ( n = 74). Quantitative reverse transcription polymerase chain reaction (RT-PCR) was used to measure GCLC messenger RNA (mRNA) expression. RESULTS Both total glutathione ( t(66) = 2.467, p = 0.016) and reduced GSH ( t(66) = 3.001, p = 0.004) levels were significantly less in people with schizophrenia than in controls. However, there were no significant differences in either GCLC-like protein ( t(72) = -1.077, p = 0.285) or GCLC mRNA expression ( t(71) = -0.376, p = 0.708) between people with schizophrenia and control subjects. There was also no significant difference of GPx-like protein levels between schizophrenia and controls ( t(72) = -0.060, p = 0.952). Moreover, no significant correlations of putative confounding factors with GSH changes were detected. DISCUSSION These results suggest that people with schizophrenia have impaired GSH antioxidant capacity, alongside normal levels of key regulatory proteins.
Collapse
Affiliation(s)
- Yiru Zhang
- 1 Schizophrenia Research Institute, Sydney, NSW, Australia.,2 Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia.,3 School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Vibeke Sørensen Catts
- 1 Schizophrenia Research Institute, Sydney, NSW, Australia.,2 Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia.,3 School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Cynthia Shannon Weickert
- 1 Schizophrenia Research Institute, Sydney, NSW, Australia.,2 Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia.,3 School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
126
|
Mohammadi A, Rashidi E, Amooeian VG. Brain, blood, cerebrospinal fluid, and serum biomarkers in schizophrenia. Psychiatry Res 2018; 265:25-38. [PMID: 29680514 DOI: 10.1016/j.psychres.2018.04.036] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/20/2018] [Accepted: 04/11/2018] [Indexed: 12/29/2022]
Abstract
Over the last decade, finding a reliable biomarker for the early detection of schizophrenia (Scz) has been a topic of interest. The main goal of the current review is to provide a comprehensive view of the brain, blood, cerebrospinal fluid (CSF), and serum biomarkers of Scz disease. Imaging studies have demonstrated that the volumes of the corpus callosum, thalamus, hippocampal formation, subiculum, parahippocampal gyrus, superior temporal gyrus, prefrontal and orbitofrontal cortices, and amygdala-hippocampal complex were reduced in patients diagnosed with Scz. It has been revealed that the levels of interleukin 1β (IL-1β), IL-6, IL-8, and TNF-α were increased in patients with Scz. Decreased mRNA levels of brain-derived neurotrophic factor (BDNF), tropomyosin receptor kinase B (TrkB), neurotrophin-3 (NT-3), nerve growth factor (NGF), and vascular endothelial growth factor (VEGF) genes have also been reported in Scz patients. Genes with known strong relationships with this disease include BDNF, catechol-O-methyltransferase (COMT), regulator of G-protein signaling 4 (RGS4), dystrobrevin-binding protein 1 (DTNBP1), neuregulin 1 (NRG1), Reelin (RELN), Selenium-binding protein 1 (SELENBP1), glutamic acid decarboxylase 67 (GAD 67), and disrupted in schizophrenia 1 (DISC1). The levels of dopamine, tyrosine hydroxylase (TH), serotonin or 5-hydroxytryptamine (5-HT) receptor 1A and B (5-HTR1A and 5-HTR1B), and 5-HT1B were significantly increased in Scz patients, while the levels of gamma-aminobutyric acid (GABA), 5-HT transporter (5-HTT), and 5-HT receptor 2A (5-HTR2A) were decreased. The increased levels of SELENBP1 and Glycogen synthase kinase 3 subunit α (GSK3α) genes in contrast with reduced levels of B-cell translocation gene 1 (BTG1), human leukocyte antigen DRB1 (HLA-DRB1), heterogeneous nuclear ribonucleoprotein A3 (HNRPA3), and serine/arginine-rich splicing factor 1 (SFRS1) genes have also been reported. This review covers various dysregulation of neurotransmitters and also highlights the strengths and weaknesses of studies attempting to identify candidate biomarkers.
Collapse
Affiliation(s)
- Alireza Mohammadi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Ehsan Rashidi
- Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Ghasem Amooeian
- Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
127
|
Transcriptome alterations of prefrontal cortical parvalbumin neurons in schizophrenia. Mol Psychiatry 2018; 23:1606-1613. [PMID: 29112193 PMCID: PMC5938166 DOI: 10.1038/mp.2017.216] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 07/14/2017] [Accepted: 08/30/2017] [Indexed: 12/19/2022]
Abstract
Schizophrenia (SZ) is associated with dysfunction of the dorsolateral prefrontal cortex (DLPFC). This dysfunction is manifest as cognitive deficits that appear to arise from disturbances in gamma frequency oscillations. These oscillations are generated in DLPFC layer 3 (L3) via reciprocal connections between pyramidal cells (PCs) and parvalbumin (PV)-containing interneurons. The density of cortical PV neurons is not altered in SZ, but expression levels of several transcripts involved in PV cell function, including PV, are lower in the disease. However, the transcriptome of PV cells has not been comprehensively assessed in a large cohort of subjects with SZ. In this study, we combined an immunohistochemical approach, laser microdissection, and microarray profiling to analyze the transcriptome of DLPFC L3 PV cells in 36 matched pairs of SZ and unaffected comparison subjects. Over 800 transcripts in PV neurons were identified as differentially expressed in SZ subjects; most of these alterations have not previously been reported. The altered transcripts were enriched for pathways involved in mitochondrial function and tight junction signaling. Comparison with the transcriptome of L3 PCs from the same subjects revealed both shared and distinct disease-related effects on gene expression between cell types. Furthermore, network structures of gene pathways differed across cell types and subject groups. These findings provide new insights into cell type-specific molecular alterations in SZ which may point toward novel strategies for identifying therapeutic targets.
Collapse
|
128
|
Avramopoulos D. Recent Advances in the Genetics of Schizophrenia. MOLECULAR NEUROPSYCHIATRY 2018; 4:35-51. [PMID: 29998117 PMCID: PMC6032037 DOI: 10.1159/000488679] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/21/2018] [Indexed: 12/27/2022]
Abstract
The last decade brought tremendous progress in the field of schizophrenia genetics. As a result of extensive collaborations and multiple technological advances, we now recognize many types of genetic variants that increase the risk. These include large copy number variants, rare coding inherited and de novο variants, and over 100 loci harboring common risk variants. While the type and contribution to the risk vary among genetic variants, there is concordance in the functions of genes they implicate, such as those whose RNA binds the fragile X-related protein FMRP and members of the activity-regulated cytoskeletal complex involved in learning and memory. Gene expression studies add important information on the biology of the disease and recapitulate the same functional gene groups. Studies of alternative phenotypes help us widen our understanding of the genetic architecture of mental function and dysfunction, how diseases overlap not only with each other but also with non-disease phenotypes. The challenge is to apply this new knowledge to prevention and treatment and help patients. The data generated so far and emerging technologies, including new methods in cell engineering, offer significant promise that in the next decade we will unlock the translational potential of these significant discoveries.
Collapse
Affiliation(s)
- Dimitrios Avramopoulos
- Institute of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Psychiatry, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
129
|
Schwede M, Nagpal S, Gandal MJ, Parikshak NN, Mirnics K, Geschwind DH, Morrow EM. Strong correlation of downregulated genes related to synaptic transmission and mitochondria in post-mortem autism cerebral cortex. J Neurodev Disord 2018; 10:18. [PMID: 29859039 PMCID: PMC5984825 DOI: 10.1186/s11689-018-9237-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 05/22/2018] [Indexed: 12/22/2022] Open
Abstract
Background Genetic studies in autism have pinpointed a heterogeneous group of loci and genes. Further, environment may be an additional factor conferring susceptibility to autism. Transcriptome studies investigate quantitative differences in gene expression between patient-derived tissues and control. These studies may pinpoint genes relevant to pathophysiology yet circumvent the need to understand genetic architecture or gene-by-environment interactions leading to disease. Methods We conducted alternate gene set enrichment analyses using differentially expressed genes from a previously published RNA-seq study of post-mortem autism cerebral cortex. We used three previously published microarray datasets for validation and one of the microarray datasets for additional differential expression analysis. The RNA-seq study used 26 autism and 33 control brains in differential gene expression analysis, and the largest microarray dataset contained 15 autism and 16 control post-mortem brains. Results While performing a gene set enrichment analysis of genes differentially expressed in the RNA-seq study, we discovered that genes associated with mitochondrial function were downregulated in autism cerebral cortex, as compared to control. These genes were correlated with genes related to synaptic function. We validated these findings across the multiple microarray datasets. We also did separate differential expression and gene set enrichment analyses to confirm the importance of the mitochondrial pathway among downregulated genes in post-mortem autism cerebral cortex. Conclusions We found that genes related to mitochondrial function were differentially expressed in autism cerebral cortex and correlated with genes related to synaptic transmission. Our principal findings replicate across all datasets investigated. Further, these findings may potentially replicate in other diseases, such as in schizophrenia. Electronic supplementary material The online version of this article (10.1186/s11689-018-9237-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Matthew Schwede
- Department of Molecular Biology, Cell Biology and Biochemistry, and Carney Institute for Brain Science, Brown University, Providence, RI, 02912, USA
| | - Shailender Nagpal
- Department of Molecular Biology, Cell Biology and Biochemistry, and Carney Institute for Brain Science, Brown University, Providence, RI, 02912, USA
| | - Michael J Gandal
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.,Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Neelroop N Parikshak
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.,Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Karoly Mirnics
- Department of Psychiatry and Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN, 37203, USA.,Present address: Department of Psychiatry, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Daniel H Geschwind
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.,Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Eric M Morrow
- Department of Molecular Biology, Cell Biology and Biochemistry, and Carney Institute for Brain Science, Brown University, Providence, RI, 02912, USA. .,Developmental Disorders Genetics Research Program, Emma Pendleton Bradley Hospital and Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, East Providence, RI, 02915, USA. .,Hassenfeld Child Health Innovation Institute, Brown University, Providence, RI, 02912, USA. .,Laboratories for Molecular Medicine, Brown University, 70 Ship Street, Box G-E4, Providence, RI, 02912, USA.
| |
Collapse
|
130
|
Cardarelli RA, Martin R, Jaaro-Peled H, Sawa A, Powell EM, O'Donnell P. Dominant-Negative DISC1 Alters the Dopaminergic Modulation of Inhibitory Interneurons in the Mouse Prefrontal Cortex. MOLECULAR NEUROPSYCHIATRY 2018; 4:20-29. [PMID: 29998115 DOI: 10.1159/000488030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 02/26/2018] [Indexed: 11/19/2022]
Abstract
A truncated disrupted in schizophrenia 1 (Disc1) gene increases the risk of psychiatric disorders, probably affecting cortical interneurons. Here, we sought to determine whether this cell population is affected in mice carrying a truncated (Disc1) allele (DN-DISC1). We utilized whole cell recordings to assess electrophysiological properties and modulation by dopamine (DA) in two classes of interneurons: fast-spiking (FS) and low threshold-spiking (LTS) interneurons in wild-type and DN-DISC1 mice. In DN-DISC1 mice, FS interneurons, but not LTS interneurons, exhibited altered action potentials. Further, the perineuronal nets that surround FS interneurons exhibited abnormal morphology in DN-DISC1 mice, and the DA modulation of this cell type was altered in DN-DISC1 mice. We conclude that early-life manipulation of a gene associated with risk of psychiatric disease can result in dysfunction, but not loss, of specific GABAergic interneurons. The resulting alteration of excitatory-inhibitory balance is a critical element in DISC1 pathophysiology.
Collapse
Affiliation(s)
- Ross A Cardarelli
- Program in Neuroscience, University of Maryland Medical School, Baltimore, Maryland, USA
| | - Rolicia Martin
- Department of Anatomy and Neurobiology, University of Maryland Medical School, Baltimore, Maryland, USA
| | - Hanna Jaaro-Peled
- Department of Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Akira Sawa
- Department of Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Elizabeth M Powell
- Department of Anatomy and Neurobiology, University of Maryland Medical School, Baltimore, Maryland, USA.,Department of Psychiatry, University of Maryland Medical School, Baltimore, Maryland, USA
| | - Patricio O'Donnell
- Program in Neuroscience, University of Maryland Medical School, Baltimore, Maryland, USA.,Department of Anatomy and Neurobiology, University of Maryland Medical School, Baltimore, Maryland, USA.,Department of Psychiatry, University of Maryland Medical School, Baltimore, Maryland, USA
| |
Collapse
|
131
|
Schulte JT, Wierenga CJ, Bruining H. Chloride transporters and GABA polarity in developmental, neurological and psychiatric conditions. Neurosci Biobehav Rev 2018; 90:260-271. [PMID: 29729285 DOI: 10.1016/j.neubiorev.2018.05.001] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/20/2018] [Accepted: 05/01/2018] [Indexed: 12/22/2022]
Abstract
Neuronal chloride regulation is a determinant factor for the dynamic tuning of GABAergic inhibition during and beyond brain development. This regulation is mainly dependent on the two co-transporters K+/Cl- co-transporter KCC2 and Na+/K+/Cl- co-transporter NKCC1, whose activity can decrease or increase neuronal chloride concentrations respectively. Altered expression and/or activity of either of these co-transporters has been associated with a wide variety of brain disorders including developmental disorders, epilepsy, schizophrenia and stroke. Here, we review current knowledge on chloride transporter expression and activity regulation and highlight the intriguing potential for existing and future interventions to support chloride homeostasis across a wide range of mental disorders and neurological conditions.
Collapse
Affiliation(s)
- Joran T Schulte
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center, Heidelberglaan 100, 3508 GA Utrecht The Netherlands
| | - Corette J Wierenga
- Division of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Hilgo Bruining
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center, Heidelberglaan 100, 3508 GA Utrecht The Netherlands.
| |
Collapse
|
132
|
Du X, Serena K, Hwang WJ, Grech A, Wu Y, Schroeder A, Hill R. Prefrontal cortical parvalbumin and somatostatin expression and cell density increase during adolescence and are modified by BDNF and sex. Mol Cell Neurosci 2018; 88:177-188. [DOI: 10.1016/j.mcn.2018.02.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 01/25/2018] [Accepted: 02/01/2018] [Indexed: 01/21/2023] Open
|
133
|
Chromosomal Conformations and Epigenomic Regulation in Schizophrenia. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 157:21-40. [PMID: 29933951 DOI: 10.1016/bs.pmbts.2017.11.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chromosomal conformations, including promoter-enhancer loops, provide a critical regulatory layer for the transcriptional machinery. Therefore, schizophrenia, a common psychiatric disorder associated with broad changes in neuronal gene expression in prefrontal cortex and other brain regions implicated in psychosis, could be associated with alterations in higher-order chromatin. Here, we review early studies on spatial genome organization in the schizophrenia postmortem brain and discuss how integrative approaches using cell culture and animal model systems could gain deeper insight into the potential roles of higher-order chromatin for the neurobiology of and novel treatment avenues for common psychiatric disease.
Collapse
|
134
|
Scarr E, Udawela M, Dean B. Changed frontal pole gene expression suggest altered interplay between neurotransmitter, developmental, and inflammatory pathways in schizophrenia. NPJ SCHIZOPHRENIA 2018; 4:4. [PMID: 29463818 PMCID: PMC5820249 DOI: 10.1038/s41537-018-0044-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 01/10/2018] [Accepted: 01/16/2018] [Indexed: 12/22/2022]
Abstract
Schizophrenia (Sz) probably occurs after genetically susceptible individuals encounter a deleterious environmental factor that triggers epigenetic mechanisms to change CNS gene expression. To determine if omnibus changes in CNS gene expression are present in Sz, we compared mRNA levels in the frontal pole (Brodmann’s area (BA) 10), the dorsolateral prefrontal cortex (BA 9) and cingulate cortex (BA 33) from 15 subjects with Sz and 15 controls using the Affymetrix™ Human Exon 1.0 ST Array. Differences in mRNA levels (±≥20%; p < 0.01) were identified (JMP Genomics 5.1) and used to predict pathways and gene x gene interactions that would be affected by the changes in gene expression using Ingenuity Pathway Analysis. There was significant variation in mRNA levels with diagnoses for 566 genes in BA 10, 65 genes in BA 9 and 40 genes in BA 33. In Sz, there was an over-representation of genes with changed expression involved in inflammation and development in BA 10, cell morphology in BA 9 and amino acid metabolism and small molecule biochemistry in BA 33. Using 94 genes with altered levels of expression in BA 10 from subjects with Sz, it was possible to construct an interactome of proven direct gene x gene interactions that was enriched for genes in inflammatory, developmental, oestrogen, serotonergic, cholinergic and NRG1 regulated pathways. Our data shows complex, regionally specific changes in cortical gene expression in Sz that are predicted to affect homeostasis between biochemical pathways already proposed to be important in the pathophysiology of the disorder. Anterior brain regions exhibit significant amounts of differentially-expressed genes which might cause dysfunction in schizophrenia. It’s thought that schizophrenia occurs when environmental factors trigger gene expression changes and downstream effects in the human brain, though this is not fully understood. An Australian research group led by Brian Dean, from the Florey Institute of Neuroscience and Mental Health, conducted a post-mortem human brain study in which they compared gene expression between 15 schizophrenia patients and 15 controls. They found 566 instances of altered gene expression in the most frontal part of the brain, Brodmann Area 10, and fewer changes in proximal regions. These are brain areas known to mediate schizophrenia-related traits and the changes in gene expression in these areas will affect a range of essential biological pathways. The group also found 97 differentially-expressed genes that have been shown to directly interact with each. This study paints a complex picture of the causes of schizophrenia but suggests modern technologies can help unravel these complexities.
Collapse
Affiliation(s)
- Elizabeth Scarr
- Molecular Psychiatry Laboratory, Florey Institute for Neuroscience and Mental Health, Parkville, VIC 3052, Australia.,CRC for Mental Health, Carlton, VIC, 3053, Australia.,Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Madhara Udawela
- Molecular Psychiatry Laboratory, Florey Institute for Neuroscience and Mental Health, Parkville, VIC 3052, Australia.,CRC for Mental Health, Carlton, VIC, 3053, Australia
| | - Brian Dean
- Molecular Psychiatry Laboratory, Florey Institute for Neuroscience and Mental Health, Parkville, VIC 3052, Australia. .,CRC for Mental Health, Carlton, VIC, 3053, Australia. .,Research Centre for Mental Health, the Faculty of Health, Arts and Design, Swinburne University, Hawthorne, VIC, 3122, Australia.
| |
Collapse
|
135
|
Charvet CJ, Šimić G, Kostović I, Knezović V, Vukšić M, Babić Leko M, Takahashi E, Sherwood CC, Wolfe MD, Finlay BL. Coevolution in the timing of GABAergic and pyramidal neuron maturation in primates. Proc Biol Sci 2018; 284:rspb.2017.1169. [PMID: 28855363 DOI: 10.1098/rspb.2017.1169] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 07/21/2017] [Indexed: 01/12/2023] Open
Abstract
The cortex of primates is relatively expanded compared with many other mammals, yet little is known about what developmental processes account for the expansion of cortical subtype numbers in primates, including humans. We asked whether GABAergic and pyramidal neuron production occurs for longer than expected in primates than in mice in a sample of 86 developing primate and rodent brains. We use high-resolution structural, diffusion MR scans and histological material to compare the timing of the ganglionic eminences (GE) and cortical proliferative pool (CPP) maturation between humans, macaques, rats, and mice. We also compare the timing of post-neurogenetic maturation of GABAergic and pyramidal neurons in primates (i.e. humans, macaques) relative to rats and mice to identify whether delays in neurogenesis are concomitant with delayed post-neurogenetic maturation. We found that the growth of the GE and CPP are both selectively delayed compared with other events in primates. By contrast, the timing of post-neurogenetic GABAergic and pyramidal events (e.g. synaptogenesis) are predictable from the timing of other events in primates and in studied rodents. The extended duration of GABAergic and pyramidal neuron production is associated with the amplification of GABAerigc and pyramidal neuron numbers in the human and non-human primate cortex.
Collapse
Affiliation(s)
- Christine J Charvet
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA .,Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC, USA
| | - Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ivica Kostović
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Vinka Knezović
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Mario Vukšić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Emi Takahashi
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Chet C Sherwood
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC, USA
| | - Marnin D Wolfe
- School of Integrative Plant Science, Department of Psychology, Cornell University, Ithaca, NY, USA
| | - Barbara L Finlay
- Evolutionary Neuroscience Group, Department of Psychology, Cornell University, Ithaca, NY, USA
| |
Collapse
|
136
|
Ovenden ES, McGregor NW, Emsley RA, Warnich L. DNA methylation and antipsychotic treatment mechanisms in schizophrenia: Progress and future directions. Prog Neuropsychopharmacol Biol Psychiatry 2018; 81:38-49. [PMID: 29017764 DOI: 10.1016/j.pnpbp.2017.10.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 10/01/2017] [Accepted: 10/04/2017] [Indexed: 12/15/2022]
Abstract
Antipsychotic response in schizophrenia is a complex, multifactorial trait influenced by pharmacogenetic factors. With genetic studies thus far providing little biological insight or clinical utility, the field of pharmacoepigenomics has emerged to tackle the so-called "missing heritability" of drug response in disease. Research on psychiatric disorders has only recently started to assess the link between epigenetic alterations and treatment outcomes. DNA methylation, the best characterised epigenetic mechanism to date, is discussed here in the context of schizophrenia and antipsychotic treatment outcomes. The majority of published studies have assessed the influence of antipsychotics on methylation levels in specific neurotransmitter-associated candidate genes or at the genome-wide level. While these studies illustrate the epigenetic modifications associated with antipsychotics, very few have assessed clinical outcomes and the potential of differential DNA methylation profiles as predictors of antipsychotic response. Results from other psychiatric disorder studies, such as depression and bipolar disorder, provide insight into what may be achieved by schizophrenia pharmacoepigenomics. Other aspects that should be addressed in future research include methodological challenges, such as tissue specificity, and the influence of genetic variation on differential methylation patterns.
Collapse
Affiliation(s)
- Ellen S Ovenden
- Department of Genetics, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Nathaniel W McGregor
- Department of Genetics, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Robin A Emsley
- Department of Psychiatry, Stellenbosch University, Tygerberg 7505, South Africa
| | - Louise Warnich
- Department of Genetics, Stellenbosch University, Stellenbosch 7600, South Africa.
| |
Collapse
|
137
|
Luoni A, Gass P, Brambilla P, Ruggeri M, Riva MA, Inta D. Altered expression of schizophrenia-related genes in mice lacking mGlu5 receptors. Eur Arch Psychiatry Clin Neurosci 2018; 268:77-87. [PMID: 27581816 DOI: 10.1007/s00406-016-0728-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 08/22/2016] [Indexed: 12/15/2022]
Abstract
The evidence underlying the so-called glutamatergic hypothesis ranges from NMDA receptor hypofunction to an imbalance between excitatory and inhibitory circuits in specific brain structures. Among all glutamatergic system components, metabotropic receptors play a main role in regulating neuronal excitability and synaptic plasticity. Here, we investigated, using qRT-PCR and western blot, consequences in the hippocampus and prefrontal/frontal cortex (PFC/FC) of mice with a genetic deletion of the metabotropic glutamate receptor 5 (mGlu5), addressing key components of the GABAergic and glutamatergic systems. We found that mGlu5 knockout (KO) mice showed a significant reduction of reelin, GAD65, GAD67 and parvalbumin mRNA levels, which is specific for the PFC/FC, and that is paralleled by a significant reduction of protein levels in male KO mice. We next analyzed the main NMDA and AMPA receptor subunits, namely GluN1, GluN2A, GluN2B and GluA1, and we found that mGlu5 deletion determined a significant reduction of their mRNA levels, also within the hippocampus, with differences between the two genders. Our data suggest that neurochemical abnormalities impinging the glutamatergic and GABAergic systems may be responsible for the behavioral phenotype associated with mGlu5 KO animals and point to the close interaction of these molecular players for the development of neuropsychiatric disorders such as schizophrenia. These data could contribute to a better understanding of the involvement of mGlu5 alterations in the molecular imbalance between excitation and inhibition underlying the emergence of a schizophrenic-like phenotype and to understand the potential of mGlu5 modulators in reversing the deficits characterizing the schizophrenic pathology.
Collapse
Affiliation(s)
- Alessia Luoni
- Department of Pharmacological and Biomolecular Sciences, Center of Neuropharmacology, Università degli Studi di Milano, Milan, Italy
| | - Peter Gass
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health Mannheim, Medical Faculty Mannheim, University of Heidelberg, J 5, 68159, Mannheim, Germany
| | - Paolo Brambilla
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Mirella Ruggeri
- Section of Psychiatry, Department of Neurological, Biomedical and Movement Sciences, University of Verona, Verona, Italy
| | - Marco A Riva
- Department of Pharmacological and Biomolecular Sciences, Center of Neuropharmacology, Università degli Studi di Milano, Milan, Italy
| | - Dragos Inta
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health Mannheim, Medical Faculty Mannheim, University of Heidelberg, J 5, 68159, Mannheim, Germany. .,Department of Psychiatry (UPK), University of Basel, Wilhelm Klein-Str. 27, 4012, Basel, Switzerland.
| |
Collapse
|
138
|
Abstract
Schizophrenia is a complex disorder lacking an effective treatment option for the pervasive and debilitating cognitive impairments experienced by patients. Working memory is a core cognitive function impaired in schizophrenia that depends upon activation of distributed neural network, including the circuitry of the dorsolateral prefrontal cortex (DLPFC). Accordingly, individuals diagnosed with schizophrenia show reduced DLPFC activation while performing working-memory tasks. This lower DLPFC activation appears to be an integral part of the disease pathophysiology, and not simply a reflection of poor performance. Thus, the cellular and circuitry alterations that underlie lower DLPFC neuronal activity in schizophrenia must be determined in order to identify appropriate therapeutic targets. Studies using human postmortem brain tissue provide a robust way to investigate and characterize these cellular and circuitry alterations at multiple levels of resolution, and such studies provide essential information that cannot be obtained either through in vivo studies in humans or through experimental animal models. Studies examining neuronal morphology, protein expression and localization, and transcript levels indicate that a microcircuit composed of excitatory pyramidal cells and inhibitory interneurons containing the calcium-binding protein parvalbumin is altered in the DLPFC of subjects with schizophrenia and likely contributes to DLPFC dysfunction.
Collapse
Affiliation(s)
- Jill R Glausier
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - David A Lewis
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
139
|
Renard J, Rushlow WJ, Laviolette SR. Effects of Adolescent THC Exposure on the Prefrontal GABAergic System: Implications for Schizophrenia-Related Psychopathology. Front Psychiatry 2018; 9:281. [PMID: 30013490 PMCID: PMC6036125 DOI: 10.3389/fpsyt.2018.00281] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/11/2018] [Indexed: 12/14/2022] Open
Abstract
Marijuana is the most commonly used drug of abuse among adolescents. Considerable clinical evidence supports the hypothesis that adolescent neurodevelopmental exposure to high levels of the principal psychoactive component in marijuana, -delta-9-tetrahydrocanabinol (THC), is associated with a high risk of developing psychiatric diseases, such as schizophrenia later in life. This marijuana-associated risk is believed to be related to increasing levels of THC found within commonly used marijuana strains. Adolescence is a highly vulnerable period for the development of the brain, where the inhibitory GABAergic system plays a pivotal role in the maturation of regulatory control mechanisms in the central nervous system (CNS). Specifically, adolescent neurodevelopment represents a critical period wherein regulatory connectivity between higher-order cortical regions and sub-cortical emotional processing circuits such as the mesolimbic dopamine (DA) system is established. Emerging preclinical evidence demonstrates that adolescent exposure to THC selectively targets schizophrenia-related molecular and neuropharmacological signaling pathways in both cortical and sub-cortical regions, including the prefrontal cortex (PFC) and mesolimbic DA pathway, comprising the ventral tegmental area (VTA) and nucleus accumbens (NAc). Prefrontal cortical GABAergic hypofunction is a key feature of schizophrenia-like neuropsychopathology. This GABAergic hypofunction may lead to the loss of control of the PFC to regulate proper sub-cortical DA neurotransmission, thereby leading to schizophrenia-like symptoms. This review summarizes preclinical evidence demonstrating that reduced prefrontal cortical GABAergic neurotransmission has a critical role in the sub-cortical DAergic dysregulation and schizophrenia-like behaviors observed following adolescent THC exposure.
Collapse
Affiliation(s)
- Justine Renard
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Walter J Rushlow
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada.,Department of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Steven R Laviolette
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada.,Department of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
140
|
The GABRB3 Polymorphism and its Association with Schizophrenia. J Mol Neurosci 2017; 64:75-79. [PMID: 29196882 DOI: 10.1007/s12031-017-1003-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 11/23/2017] [Indexed: 10/18/2022]
Abstract
The aim of this study was to explore whether schizophrenia occurrence is associated with polymorphisms in the 5' regulatory region of GABRB3 (gamma-aminobutyric acid type A receptor beta 3, subunit gene). The study included 324 patients with schizophrenia and 327 unaffected participants; all individuals were northern Han Chinese. Genotype and haplotype frequency distributions were compared for the 2 groups by means of PCR amplification and direct sequencing of the promoter region of GABRB3. The genotype distribution among control participants was in accordance with the Hardy-Weinberg equilibrium. Five common single-nucleotide polymorphism (SNP) sites were detected in the 5' promoter region of GABRB3: rs4243768, rs7171660, rs4363842, rs4906902, and rs8179184. Only rs8179184 and rs4906902 differed significantly in frequency between controls and cases (P < 0.05); this difference remained significant when only women in each group were compared. The 2 SNP sites showed linkage disequilibrium, resulting in 2 haplotypes: T-G and C-A. The frequency of C-A was significantly higher among patients with schizophrenia than among controls. Our findings suggest that rs4906902 and rs8179184 in the 5' promoter region of GABRB3 are associated with schizophrenia. The C-A haplotype may entail an increased risk of schizophrenia, and the onset of schizophrenia may be gender-specific.
Collapse
|
141
|
Challenges and opportunities for the development of new antipsychotic drugs. Biochem Pharmacol 2017; 143:10-24. [DOI: 10.1016/j.bcp.2017.05.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 05/12/2017] [Indexed: 12/20/2022]
|
142
|
Zhu J, Zhuo C, Xu L, Liu F, Qin W, Yu C. Altered Coupling Between Resting-State Cerebral Blood Flow and Functional Connectivity in Schizophrenia. Schizophr Bull 2017; 43:1363-1374. [PMID: 28521048 PMCID: PMC5737873 DOI: 10.1093/schbul/sbx051] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Respective changes in resting-state cerebral blood flow (CBF) and functional connectivity in schizophrenia have been reported. However, their coupling alterations in schizophrenia remain largely unknown. METHODS 89 schizophrenia patients and 90 sex- and age-matched healthy controls underwent resting-state functional MRI to calculate functional connectivity strength (FCS) and arterial spin labeling imaging to compute CBF. The CBF-FCS coupling of the whole gray matter and the CBF/FCS ratio (the amount of blood supply per unit of connectivity strength) of each voxel were compared between the 2 groups. RESULTS Whole gray matter CBF-FCS coupling was decreased in schizophrenia patients relative to healthy controls. In schizophrenia patients, the decreased CBF/FCS ratio was predominantly located in cognitive- and emotional-related brain regions, including the dorsolateral prefrontal cortex, insula, hippocampus and thalamus, whereas an increased CBF/FCS ratio was mainly identified in the sensorimotor regions, including the putamen, and sensorimotor, mid-cingulate and visual cortices. CONCLUSION These findings suggest that the neurovascular decoupling in the brain may be a possible neuropathological mechanism of schizophrenia.
Collapse
Affiliation(s)
- Jiajia Zhu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Chuanjun Zhuo
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China,Department of Psychiatry Functional Neuroimaging Laboratory, Tianjin Mental Health Center, Tianjin Anding Hospital, Tianjin, China,Tianjin Anning Hospital, Tianjin, China
| | - Lixue Xu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Feng Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Wen Qin
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Chunshui Yu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China,To whom correspondence should be addressed; Department of Radiology, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China; tel: +86-22-63062026, fax: +86-22-63062290, e-mail:
| |
Collapse
|
143
|
Renard J, Szkudlarek HJ, Kramar CP, Jobson CEL, Moura K, Rushlow WJ, Laviolette SR. Adolescent THC Exposure Causes Enduring Prefrontal Cortical Disruption of GABAergic Inhibition and Dysregulation of Sub-Cortical Dopamine Function. Sci Rep 2017; 7:11420. [PMID: 28900286 PMCID: PMC5595795 DOI: 10.1038/s41598-017-11645-8] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 08/29/2017] [Indexed: 02/07/2023] Open
Abstract
Chronic adolescent marijuana use has been linked to the later development of psychiatric diseases such as schizophrenia. GABAergic hypofunction in the prefrontal cortex (PFC) is a cardinal pathological feature of schizophrenia and may be a mechanism by which the PFC loses its ability to regulate sub-cortical dopamine (DA) resulting in schizophrenia-like neuropsychopathology. In the present study, we exposed adolescent rats to Δ-9-tetra-hydrocannabinol (THC), the psychoactive component in marijuana. At adulthood, we characterized the functionality of PFC GABAergic neurotransmission and its regulation of sub-cortical DA function using molecular, behavioral and in-vivo electrophysiological analyses. Our findings revealed a persistent attenuation of PFC GABAergic function combined with a hyperactive neuronal state in PFC neurons and associated disruptions in cortical gamma oscillatory activity. These PFC abnormalities were accompanied by hyperactive DAergic neuronal activity in the ventral tegmental area (VTA) and behavioral and cognitive abnormalities similar to those observed in psychiatric disorders. Remarkably, these neuronal and behavioral effects were reversed by pharmacological activation of GABAA receptors in the PFC. Together, these results identify a mechanistic link between dysregulated frontal cortical GABAergic inhibition and sub-cortical DAergic dysregulation, characteristic of well-established neuropsychiatric endophenotypes.
Collapse
Affiliation(s)
- Justine Renard
- Dept. of Anatomy and Cell Biology & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Hanna J Szkudlarek
- Dept. of Anatomy and Cell Biology & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Cecilia P Kramar
- Dept. of Anatomy and Cell Biology & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Christina E L Jobson
- Dept. of Anatomy and Cell Biology & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Kyra Moura
- Dept. of Anatomy and Cell Biology & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Walter J Rushlow
- Dept. of Anatomy and Cell Biology & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada.,Dept. of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Steven R Laviolette
- Dept. of Anatomy and Cell Biology & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada. .,Dept. of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada.
| |
Collapse
|
144
|
Batista-Brito R, Vinck M, Ferguson KA, Chang JT, Laubender D, Lur G, Mossner JM, Hernandez VG, Ramakrishnan C, Deisseroth K, Higley MJ, Cardin JA. Developmental Dysfunction of VIP Interneurons Impairs Cortical Circuits. Neuron 2017; 95:884-895.e9. [PMID: 28817803 DOI: 10.1016/j.neuron.2017.07.034] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 07/08/2017] [Accepted: 07/27/2017] [Indexed: 02/01/2023]
Abstract
GABAergic interneurons play important roles in cortical circuit development. However, there are multiple populations of interneurons and their respective developmental contributions remain poorly explored. Neuregulin 1 (NRG1) and its interneuron-specific receptor ERBB4 are critical genes for interneuron maturation. Using a conditional ErbB4 deletion, we tested the role of vasoactive intestinal peptide (VIP)-expressing interneurons in the postnatal maturation of cortical circuits in vivo. ErbB4 removal from VIP interneurons during development leads to changes in their activity, along with severe dysregulation of cortical temporal organization and state dependence. These alterations emerge during adolescence, and mature animals in which VIP interneurons lack ErbB4 exhibit reduced cortical responses to sensory stimuli and impaired sensory learning. Our data support a key role for VIP interneurons in cortical circuit development and suggest a possible contribution to pathophysiology in neurodevelopmental disorders. These findings provide a new perspective on the role of GABAergic interneuron diversity in cortical development. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Renata Batista-Brito
- Yale University School of Medicine, Department of Neuroscience, 333 Cedar St., New Haven, CT, 06520, USA; Kavli Institute of Neuroscience, Yale University, 333 Cedar St., New Haven CT, 06520, USA
| | - Martin Vinck
- Yale University School of Medicine, Department of Neuroscience, 333 Cedar St., New Haven, CT, 06520, USA; Kavli Institute of Neuroscience, Yale University, 333 Cedar St., New Haven CT, 06520, USA; Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, Deutschordenstraße 46, 60528 Frankfurt, Germany
| | - Katie A Ferguson
- Yale University School of Medicine, Department of Neuroscience, 333 Cedar St., New Haven, CT, 06520, USA; Kavli Institute of Neuroscience, Yale University, 333 Cedar St., New Haven CT, 06520, USA
| | - Jeremy T Chang
- Yale University School of Medicine, Department of Neuroscience, 333 Cedar St., New Haven, CT, 06520, USA; Kavli Institute of Neuroscience, Yale University, 333 Cedar St., New Haven CT, 06520, USA
| | - David Laubender
- Yale University School of Medicine, Department of Neuroscience, 333 Cedar St., New Haven, CT, 06520, USA; Kavli Institute of Neuroscience, Yale University, 333 Cedar St., New Haven CT, 06520, USA
| | - Gyorgy Lur
- Yale University School of Medicine, Department of Neuroscience, 333 Cedar St., New Haven, CT, 06520, USA; Kavli Institute of Neuroscience, Yale University, 333 Cedar St., New Haven CT, 06520, USA
| | - James M Mossner
- Yale University School of Medicine, Department of Neuroscience, 333 Cedar St., New Haven, CT, 06520, USA; Kavli Institute of Neuroscience, Yale University, 333 Cedar St., New Haven CT, 06520, USA
| | - Victoria G Hernandez
- Yale University School of Medicine, Department of Neuroscience, 333 Cedar St., New Haven, CT, 06520, USA; Kavli Institute of Neuroscience, Yale University, 333 Cedar St., New Haven CT, 06520, USA
| | - Charu Ramakrishnan
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; HHMI, Stanford University, Stanford, CA 94305, USA; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Michael J Higley
- Yale University School of Medicine, Department of Neuroscience, 333 Cedar St., New Haven, CT, 06520, USA; Kavli Institute of Neuroscience, Yale University, 333 Cedar St., New Haven CT, 06520, USA
| | - Jessica A Cardin
- Yale University School of Medicine, Department of Neuroscience, 333 Cedar St., New Haven, CT, 06520, USA; Kavli Institute of Neuroscience, Yale University, 333 Cedar St., New Haven CT, 06520, USA.
| |
Collapse
|
145
|
Ferreri F, Guerra A, Vollero L, Ponzo D, Maatta S, Mervaala E, Iannello G, Di Lazzaro V. Age-related changes of cortical excitability and connectivity in healthy humans: non-invasive evaluation of sensorimotor network by means of TMS-EEG. Neuroscience 2017. [DOI: 10.1016/j.neuroscience.2017.06.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Florinda Ferreri
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, University Campus Bio-Medico, via Álvaro del Portillo 21, 00128 Rome, Italy; Department of Clinical Neurophysiology, Kuopio University Hospital, University of Eastern Finland, 70029 KYS, Kuopio, Finland.
| | - Andrea Guerra
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, University Campus Bio-Medico, via Álvaro del Portillo 21, 00128 Rome, Italy
| | - Luca Vollero
- Department of Computer Science and Computer Engineering, University Campus Bio-Medico, via Álvaro del Portillo 200, 00128 Rome, Italy
| | - David Ponzo
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, University Campus Bio-Medico, via Álvaro del Portillo 21, 00128 Rome, Italy
| | - Sara Maatta
- Department of Clinical Neurophysiology, Kuopio University Hospital, University of Eastern Finland, 70029 KYS, Kuopio, Finland
| | - Esa Mervaala
- Department of Clinical Neurophysiology, Kuopio University Hospital, University of Eastern Finland, 70029 KYS, Kuopio, Finland
| | - Giulio Iannello
- Department of Computer Science and Computer Engineering, University Campus Bio-Medico, via Álvaro del Portillo 200, 00128 Rome, Italy
| | - Vincenzo Di Lazzaro
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, University Campus Bio-Medico, via Álvaro del Portillo 21, 00128 Rome, Italy
| |
Collapse
|
146
|
Chang X, Liu Y, Hahn CG, Gur RE, Sleiman PMA, Hakonarson H. RNA-seq analysis of amygdala tissue reveals characteristic expression profiles in schizophrenia. Transl Psychiatry 2017; 7:e1203. [PMID: 28809853 PMCID: PMC5611723 DOI: 10.1038/tp.2017.154] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/02/2017] [Accepted: 05/30/2017] [Indexed: 12/15/2022] Open
Abstract
The amygdala brain region has been implicated in the pathophysiology of schizophrenia through emotion processing. However, transcriptome messages in the amygdala of schizophrenia patients have not been well studied. We used RNA sequencing to investigate gene-expression profiling in the amygdala tissues, and identified 569 upregulated and 192 downregulated genes from 22 schizophrenia patients and 24 non-psychiatric controls. Gene functional enrichment analysis demonstrated that the downregulated genes were enriched in pathways such as 'synaptic transmission' and 'behavior', whereas the upregulated genes were significantly over-represented in gene ontology pathways such as 'immune response' and 'blood vessel development'. Co-expression-based gene network analysis identified seven modules including four modules significantly associated with 'synaptic transmission', 'blood vessel development' or 'immune responses'. Taken together, our study provides novel insights into the molecular mechanism of schizophrenia, suggesting that precision-tailored therapeutic approaches aimed at normalizing the expression/function of specific gene networks could be a promising option in schizophrenia.
Collapse
Affiliation(s)
- X Chang
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Y Liu
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - C-G Hahn
- Neuropsychiatric Signaling Program, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - R E Gur
- Neuropsychiatry Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - P M A Sleiman
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA,Division of Human Genetics, Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - H Hakonarson
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA,Division of Human Genetics, Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Leonard Madlyn Abramson Research Center, 3615 Civic Center Boulevard, Room 1216E, Philadelphia, PA 19104-4318, USA. E-mail:
| |
Collapse
|
147
|
Dorsolateral Prefrontal Cortex GABA Concentration in Humans Predicts Working Memory Load Processing Capacity. J Neurosci 2017; 36:11788-11794. [PMID: 27852785 DOI: 10.1523/jneurosci.1970-16.2016] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/24/2016] [Accepted: 09/19/2016] [Indexed: 02/01/2023] Open
Abstract
The discovery of neural mechanisms of working memory (WM) would significantly enhance our understanding of complex human behaviors and guide treatment development for WM-related impairments found in neuropsychiatric conditions and aging. Although the dorsolateral prefrontal cortex (DLPFC) has long been considered critical for WM, we still know little about the neural elements and pathways within the DLPFC that support WM in humans. In this study, we tested whether an individual's DLPFC gamma-aminobutryic acid (GABA) content predicts individual differences in WM task performance using a novel behavioral approach. Twenty-three healthy adults completed a task that measured the unique contribution of major WM components (memory load, maintenance, and distraction resistance) to performance. This was done to address the possibility that components have differing GABA dependencies and the failure to parse WM into components would lead to missing true associations with GABA. The subjects then had their DLPFC GABA content measured by single-voxel proton magnetic spectroscopy. We found that individuals with lower DLPFC GABA showed greater performance degradation with higher load, accounting for 31% of variance, p(corrected) = 0.015. This relationship was component, neurochemical, and brain region specific. DLPFC GABA content did not predict performance sensitivity to other components tested; DLPFC glutamate + glutamine and visual cortical GABA content did not predict load sensitivity. These results confirm the involvement of DLPFC GABA in WM load processing in humans and implicate factors controlling DLPFC GABA content in the neural mechanisms of WM and its impairments. SIGNIFICANCE STATEMENT This study demonstrated for the first time that the amount of gamma-aminobutryic acid (GABA), the major inhibitory neurotransmitter of the brain, in an individual's prefrontal cortex predicts working memory (WM) task performance. Given that WM is required for many of the most characteristic cognitive and behavioral capabilities in humans, this finding could have a significant impact on our understanding of the neural basis of complex human behavior. Furthermore, this finding suggests that efforts to preserve or increase brain GABA levels could be fruitful in remediating WM-related deficits associated with neuropsychiatric conditions.
Collapse
|
148
|
Wearne TA, Parker LM, Franklin JL, Goodchild AK, Cornish JL. Behavioral sensitization to methamphetamine induces specific interneuronal mRNA pathology across the prelimbic and orbitofrontal cortices. Prog Neuropsychopharmacol Biol Psychiatry 2017; 77:42-48. [PMID: 28351548 DOI: 10.1016/j.pnpbp.2017.03.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 03/15/2017] [Accepted: 03/23/2017] [Indexed: 12/31/2022]
Abstract
Schizophrenia is associated with significant pathophysiological changes to interneurons within the prefrontal cortex (PFC), with mRNA and protein changes associated with the GABA network localized to specific interneuron subtypes. Methamphetamine is a commonly abused psychostimulant that can induce chronic psychosis and symptoms that are similar to schizophrenia, suggesting that chronic METH induced psychosis may be associated with similar brain pathology to schizophrenia in the PFC. The aim of this study, therefore, was to examine mRNA expression of interneuron markers across two regions of the PFC (prelimbic (PRL) and orbitofrontal cortices (OFC)) following METH sensitization, an animal model of METH psychosis. We also studied the association between GABA mRNA expression and interneuronal mRNA expression to identify whether particular changes to the GABA network could be localized to a specific inhibitory cellular phenotype. METH sensitization increased the transcriptional expression of calbindin, calretinin, somatostatin, cholecyctokinin and vasoactive intestinal peptide in the PRL while parvalbumin, calbindin, cholectokinin and vasoactive intestinal peptide were upregulated in the OFC. Based on our previous findings, we also found significant correlations between GAD67, GAT1 and parvalbumin while GAD67, GAD65 and GAT1 were positively correlated with cholecystokinin in the PRL of METH sensitized rats. Within the OFC, the expression of GABAAα1 was positively correlated with somatostatin while GABAAα5 was negatively associated with somatostatin and calbindin. These findings suggest that METH sensitization differentially changes the expression of mRNAs encoding for multiple peptides and calcium binding proteins across the PRL and the OFC. Furthermore, these findings support that changes to the GABA network may also occur within specific cell types. These results, therefore, provide the first evidence that METH sensitization mediates differential interneuronal pathology across the PRL and OFC and such changes could have profound consequences on behavior and cognitive output.
Collapse
Affiliation(s)
- Travis A Wearne
- Department of Psychology, Faculty of Human Sciences, Centre for Emotional Health, Macquarie University, Sydney, NSW, Australia
| | - Lindsay M Parker
- Department of Biomedical Science, Faculty of Medicine and Health Science, Macquarie University, Sydney, NSW, Australia; ARC Center of Excellence for Nanoscale BioPhotonics, Macquarie University, Sydney, NSW, Australia
| | - Jane L Franklin
- Department of Psychology, Faculty of Human Sciences, Centre for Emotional Health, Macquarie University, Sydney, NSW, Australia
| | - Ann K Goodchild
- Department of Biomedical Science, Faculty of Medicine and Health Science, Macquarie University, Sydney, NSW, Australia
| | - Jennifer L Cornish
- Department of Psychology, Faculty of Human Sciences, Centre for Emotional Health, Macquarie University, Sydney, NSW, Australia; ARC Center of Excellence for Nanoscale BioPhotonics, Macquarie University, Sydney, NSW, Australia.
| |
Collapse
|
149
|
Balan S, Yamada K, Iwayama Y, Hashimoto T, Toyota T, Shimamoto C, Maekawa M, Takagai S, Wakuda T, Kameno Y, Kurita D, Yamada K, Kikuchi M, Hashimoto T, Kanahara N, Yoshikawa T. Comprehensive association analysis of 27 genes from the GABAergic system in Japanese individuals affected with schizophrenia. Schizophr Res 2017; 185:33-40. [PMID: 28073605 DOI: 10.1016/j.schres.2017.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 12/26/2016] [Accepted: 01/01/2017] [Indexed: 01/01/2023]
Abstract
Involvement of the gamma-aminobutyric acid (GABA)-ergic system in schizophrenia pathogenesis through disrupted neurodevelopment has been highlighted in numerous studies. However, the function of common genetic variants of this system in determining schizophrenia risk is unknown. We therefore tested the association of 375 tagged SNPs in genes derived from the GABAergic system, such as GABAA receptor subunit genes, and GABA related genes (glutamate decarboxylase genes, GABAergic-marker gene, genes involved in GABA receptor trafficking and scaffolding) in Japanese schizophrenia case-control samples (n=2926; 1415 cases and 1511 controls). We observed nominal association of SNPs in nine GABAA receptor subunit genes and the GPHN gene with schizophrenia, although none survived correction for study-wide multiple testing. Two SNPs located in the GABRA1 gene, rs4263535 (Pallele=0.002; uncorrected) and rs1157122 (Pallele=0.006; uncorrected) showed top hits, followed by rs723432 (Pallele=0.007; uncorrected) in the GPHN gene. All three were significantly associated with schizophrenia and survived gene-wide multiple testing. Haplotypes containing associated variants in GABRA1 but not GPHN were significantly associated with schizophrenia. To conclude, we provided substantiating genetic evidence for the involvement of the GABAergic system in schizophrenia susceptibility. These results warrant further investigations to replicate the association of GABRA1 and GPHN with schizophrenia and to discern the precise mechanisms of disease pathophysiology.
Collapse
Affiliation(s)
- Shabeesh Balan
- Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama 351-0198, Japan
| | - Kazuo Yamada
- Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama 351-0198, Japan
| | - Yoshimi Iwayama
- Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama 351-0198, Japan
| | - Takanori Hashimoto
- Department of Psychiatry and Neurobiology, Kanazawa University Graduate School of Medicine, Kanazawa 920-8641, Japan
| | - Tomoko Toyota
- Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama 351-0198, Japan
| | - Chie Shimamoto
- Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama 351-0198, Japan
| | - Motoko Maekawa
- Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama 351-0198, Japan
| | - Shu Takagai
- Department of Psychiatry, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Tomoyasu Wakuda
- Department of Psychiatry, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Yosuke Kameno
- Department of Psychiatry, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Daisuke Kurita
- Department of Psychiatry, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Kohei Yamada
- Department of Psychiatry, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Mitsuru Kikuchi
- Department of Psychiatry and Neurobiology, Kanazawa University Graduate School of Medicine, Kanazawa 920-8641, Japan
| | - Tasuku Hashimoto
- Department of Psychiatry, Graduate School of Medicine, Chiba University, Chiba 260-8677, Japan
| | - Nobuhisa Kanahara
- Department of Psychiatry, Graduate School of Medicine, Chiba University, Chiba 260-8677, Japan
| | - Takeo Yoshikawa
- Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama 351-0198, Japan.
| |
Collapse
|
150
|
Rocco BR, DeDionisio AM, Lewis DA, Fish KN. Alterations in a Unique Class of Cortical Chandelier Cell Axon Cartridges in Schizophrenia. Biol Psychiatry 2017; 82:40-48. [PMID: 27884423 PMCID: PMC5374057 DOI: 10.1016/j.biopsych.2016.09.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 09/24/2016] [Accepted: 09/28/2016] [Indexed: 12/28/2022]
Abstract
BACKGROUND The axons of chandelier cells (ChCs) target the axon initial segment of pyramidal neurons, forming an array of boutons termed a cartridge. In schizophrenia, the density of cartridges detectable by gamma-aminobutyric acid (GABA) membrane transporter 1 immunoreactivity is lower, whereas the density of axon initial segments detectable by immunoreactivity for the α2 subunit of the GABAA receptor is higher in layers 2/superficial 3 of the prefrontal cortex. These findings were interpreted as compensatory responses to lower GABA levels in ChCs. However, we recently found that in schizophrenia, ChC cartridge boutons contain normal levels of the 67 kDa isoform of glutamic acid decarboxylase (GAD67) protein, the enzyme responsible for GABA synthesis in these boutons. To understand these findings we quantified the densities of ChC cartridges immunoreactive for vesicular GABA transporter (vGAT+), which is present in all cartridge boutons, and the subset of cartridges that contain calbindin (CB+). METHODS Prefrontal cortex tissue sections from 20 matched pairs of schizophrenia and unaffected comparison subjects were immunolabeled for vGAT, GAD67, and CB. RESULTS The mean density of vGAT+/CB+ cartridges was 2.7-fold higher, exclusively in layer 2 of schizophrenia subjects, whereas the density of vGAT+/CB- cartridges did not differ between subject groups. Neither vGAT, CB, or GAD67 protein levels per ChC bouton nor the number of boutons per cartridge differed between subject groups. CONCLUSIONS Our findings of a greater density of CB+ ChC cartridges in prefrontal cortex layer 2 from schizophrenia subjects suggests that the normal developmental pruning of these cartridges is blunted in the illness.
Collapse
Affiliation(s)
- Brad R Rocco
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Adam M DeDionisio
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kenneth N Fish
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| |
Collapse
|