1
|
Oliveras I, Soria-Ruiz OJ, Sampedro-Viana D, Cañete T, Río-Álamos C, Tobeña A, Fernández-Teruel A. Different maturation patterns for sensorimotor gating and startle habituation deficits in male and female RHA vs RLA rats. Behav Brain Res 2022; 434:114021. [PMID: 35872331 DOI: 10.1016/j.bbr.2022.114021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 05/17/2022] [Accepted: 07/19/2022] [Indexed: 11/28/2022]
Abstract
Neurodevelopmental anomalies are thought to play a crucial role in the emergence of schizophrenia. The Roman high-avoidance (RHA) rats exhibit impaired prepulse inhibition (PPI), as well as other behavioral and cognitive singularities related to schizophrenia syndromes compared to the Roman low-avoidance (RLA) rats. In the present study, we aimed at elucidating whether PPI deficits in the RHA rats take place during prepubescence, adolescence, or adulthood. Thus, we evaluated the levels of PPI of both strains and both sexes during these three developmental phases. Additionally, we also investigated the onset of startle habituation deficits in the same groups. The results showed that male RHA rats exhibit a clear-cut PPI reduction compared to their RLA counterparts in adulthood. In female RHA rats, we observed lower levels of PPI since adolescence and through adulthood. We also found no differences between PPI percentages among the three ages in RHA male rats. Contrarily, in male RLA rats, PPI levels were increased in adults compared to their adolescent and prepubescent counterparts. Finally, a deficit in startle habituation was observed in adulthood of both male and female RHA rats, although in the latter case the disturbance in startle habituation was more profound. These results further the description of the maturational trajectory of cognitive markers relevant to schizophrenia prodrome and they add face validity to the RHA rats as a model of schizophrenia-relevant phenotypes.
Collapse
Affiliation(s)
- Ignasi Oliveras
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine & Institute of Neurosciences, School of Medicine, Autonomous University of Barcelona, Bellaterra, Barcelona 08193, Spain.
| | - Oscar J Soria-Ruiz
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine & Institute of Neurosciences, School of Medicine, Autonomous University of Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Daniel Sampedro-Viana
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine & Institute of Neurosciences, School of Medicine, Autonomous University of Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Toni Cañete
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine & Institute of Neurosciences, School of Medicine, Autonomous University of Barcelona, Bellaterra, Barcelona 08193, Spain
| | | | - Adolf Tobeña
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine & Institute of Neurosciences, School of Medicine, Autonomous University of Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Alberto Fernández-Teruel
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine & Institute of Neurosciences, School of Medicine, Autonomous University of Barcelona, Bellaterra, Barcelona 08193, Spain.
| |
Collapse
|
2
|
Receptor-Mediated AKT/PI3K Signalling and Behavioural Alterations in Zebrafish Larvae Reveal Association between Schizophrenia and Opioid Use Disorder. Int J Mol Sci 2022; 23:ijms23094715. [PMID: 35563106 PMCID: PMC9104710 DOI: 10.3390/ijms23094715] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 02/01/2023] Open
Abstract
The link between substance abuse and the development of schizophrenia remains elusive. In this study, we assessed the molecular and behavioural alterations associated with schizophrenia, opioid addiction, and opioid withdrawal using zebrafish as a biological model. Larvae of 2 days post fertilization (dpf) were exposed to domperidone (DMP), a dopamine-D2 dopamine D2 receptor antagonist, and morphine for 3 days and 10 days, respectively. MK801, an N-methyl-D-aspartate (NMDA) receptor antagonist, served as a positive control to mimic schizophrenia-like behaviour. The withdrawal syndrome was assessed 5 days after the termination of morphine treatment. The expressions of schizophrenia susceptibility genes, i.e., pi3k, akt1, slc6a4, creb1 and adamts2, in brains were quantified, and the levels of whole-body cyclic adenosine monophosphate (cAMP), serotonin and cortisol were measured. The aggressiveness of larvae was observed using the mirror biting test. After the short-term treatment with DMP and morphine, all studied genes were not differentially expressed. As for the long-term exposure, akt1 was downregulated by DMP and morphine. Downregulation of pi3k and slc6a4 was observed in the morphine-treated larvae, whereas creb1 and adamts2 were upregulated by DMP. The levels of cAMP and cortisol were elevated after 3 days, whereas significant increases were observed in all of the biochemical tests after 10 days. Compared to controls, increased aggression was observed in the DMP-, but not morphine-, treated group. These two groups showed reduction in aggressiveness when drug exposure was prolonged. Both the short- and long-term morphine withdrawal groups showed downregulation in all genes examined except creb1, suggesting dysregulated reward circuitry function. These results suggest that biochemical and behavioural alterations in schizophrenia-like symptoms and opioid dependence could be controlled by common mechanisms.
Collapse
|
3
|
Cano JC, Huang W, Fénelon K. The amygdala modulates prepulse inhibition of the auditory startle reflex through excitatory inputs to the caudal pontine reticular nucleus. BMC Biol 2021; 19:116. [PMID: 34082731 PMCID: PMC8176709 DOI: 10.1186/s12915-021-01050-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 05/12/2021] [Indexed: 01/20/2023] Open
Abstract
Background Sensorimotor gating is a fundamental pre-attentive process that is defined as the inhibition of a motor response by a sensory event. Sensorimotor gating, commonly measured using the prepulse inhibition (PPI) of the auditory startle reflex task, is impaired in patients suffering from various neurological and psychiatric disorders. PPI deficits are a hallmark of schizophrenia, and they are often associated with attention and other cognitive impairments. Although the reversal of PPI deficits in animal models is widely used in pre-clinical research for antipsychotic drug screening, the neurotransmitter systems and synaptic mechanisms underlying PPI are still not resolved, even under physiological conditions. Recent evidence ruled out the longstanding hypothesis that PPI is mediated by midbrain cholinergic inputs to the caudal pontine reticular nucleus (PnC). Instead, glutamatergic, glycinergic, and GABAergic inhibitory mechanisms are now suggested to be crucial for PPI, at the PnC level. Since amygdalar dysfunctions alter PPI and are common to pathologies displaying sensorimotor gating deficits, the present study was designed to test that direct projections to the PnC originating from the amygdala contribute to PPI. Results Using wild type and transgenic mice expressing eGFP under the control of the glycine transporter type 2 promoter (GlyT2-eGFP mice), we first employed tract-tracing, morphological reconstructions, and immunohistochemical analyses to demonstrate that the central nucleus of the amygdala (CeA) sends glutamatergic inputs lateroventrally to PnC neurons, including GlyT2+ cells. Then, we showed the contribution of the CeA-PnC excitatory synapses to PPI in vivo by demonstrating that optogenetic inhibition of this connection decreases PPI, and optogenetic activation induces partial PPI. Finally, in GlyT2-Cre mice, whole-cell recordings of GlyT2+ PnC neurons in vitro paired with optogenetic stimulation of CeA fibers, as well as photo-inhibition of GlyT2+ PnC neurons in vivo, allowed us to implicate GlyT2+ neurons in the PPI pathway. Conclusions Our results uncover a feedforward inhibitory mechanism within the brainstem startle circuit by which amygdalar glutamatergic inputs and GlyT2+ PnC neurons contribute to PPI. We are providing new insights to the clinically relevant theoretical construct of PPI, which is disrupted in various neuropsychiatric and neurological diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01050-z.
Collapse
Affiliation(s)
- Jose Carlos Cano
- Department of Biological Sciences, University of Texas at El Paso, 500 West University Avenue, El Paso, TX, 79912, USA
| | - Wanyun Huang
- Biology Department, University of Massachusetts Amherst, Life Science Laboratories, 240 Thatcher Road, Amherst, MA, 01002, USA
| | - Karine Fénelon
- Biology Department, University of Massachusetts Amherst, Life Science Laboratories, 240 Thatcher Road, Amherst, MA, 01002, USA.
| |
Collapse
|
4
|
Bühler A, Carl M. Zebrafish Tools for Deciphering Habenular Network-Linked Mental Disorders. Biomolecules 2021; 11:biom11020324. [PMID: 33672636 PMCID: PMC7924194 DOI: 10.3390/biom11020324] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/12/2021] [Accepted: 02/14/2021] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Everything that we think, feel or do depends on the function of neural networks in the brain. These are highly complex structures made of cells (neurons) and their interconnections (axons), which develop dependent on precisely coordinated interactions of genes. Any gene mutation can result in unwanted alterations in neural network formation and concomitant brain disorders. The habenula neural network is one of these important circuits, which has been linked to autism, schizophrenia, depression and bipolar disorder. Studies using the zebrafish have uncovered genes involved in the development of this network. Intriguingly, some of these genes have also been identified as risk genes of human brain disorders highlighting the power of this animal model to link risk genes and the affected network to human disease. But can we use the advantages of this model to identify new targets and compounds with ameliorating effects on brain dysfunction? In this review, we summarise the current knowledge on techniques to manipulate the habenula neural network to study the consequences on behavior. Moreover, we give an overview of existing behavioral test to mimic aspects of mental disorders and critically discuss the applicability of the zebrafish model in this field of research. Abstract The prevalence of patients suffering from mental disorders is substantially increasing in recent years and represents a major burden to society. The underlying causes and neuronal circuits affected are complex and difficult to unravel. Frequent disorders such as depression, schizophrenia, autism, and bipolar disorder share links to the habenular neural circuit. This conserved neurotransmitter system relays cognitive information between different brain areas steering behaviors ranging from fear and anxiety to reward, sleep, and social behaviors. Advances in the field using the zebrafish model organism have uncovered major genetic mechanisms underlying the formation of the habenular neural circuit. Some of the identified genes involved in regulating Wnt/beta-catenin signaling have previously been suggested as risk genes of human mental disorders. Hence, these studies on habenular genetics contribute to a better understanding of brain diseases. We are here summarizing how the gained knowledge on the mechanisms underlying habenular neural circuit development can be used to introduce defined manipulations into the system to study the functional behavioral consequences. We further give an overview of existing behavior assays to address phenotypes related to mental disorders and critically discuss the power but also the limits of the zebrafish model for identifying suitable targets to develop therapies.
Collapse
Affiliation(s)
- Anja Bühler
- Correspondence: (A.B.); (M.C.); Tel.: +39-0461-282745 (A.B.); +39-0461-283931 (M.C.)
| | - Matthias Carl
- Correspondence: (A.B.); (M.C.); Tel.: +39-0461-282745 (A.B.); +39-0461-283931 (M.C.)
| |
Collapse
|
5
|
San-Martin R, Castro LA, Menezes PR, Fraga FJ, Simões PW, Salum C. Meta-Analysis of Sensorimotor Gating Deficits in Patients With Schizophrenia Evaluated by Prepulse Inhibition Test. Schizophr Bull 2020; 46:1482-1497. [PMID: 32506125 PMCID: PMC8061122 DOI: 10.1093/schbul/sbaa059] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Prepulse inhibition (PPI) of startle is an operational measure of sensorimotor gating that is often impaired in patients with schizophrenia. Despite the large number of studies, there is considerable variation in PPI outcomes reported. We conducted a systematic review and meta-analysis investigating PPI impairment in patients with schizophrenia compared with healthy control subjects, and examined possible explanations for the variation in results between studies. Major databases were screened for observational studies comparing healthy subjects and patients with schizophrenia for the prepulse and pulse intervals of 60 and 120 ms as primary outcomes, ie, PPI-60 and PPI-120. Standardized mean difference (SMD) and 95% confidence intervals (CI) were extracted and pooled using random effects models. We then estimated the mean effect size of these measures with random effects meta-analyses and evaluated potential PPI heterogeneity moderators, using sensitivity analysis and meta-regressions. Sixty-seven primary studies were identified, with 3685 healthy and 4290 patients with schizophrenia. The schizophrenia group showed reduction in sensorimotor gating for both PPI-60 (SMD = -0.50, 95% CI = [-0.61, -0.39]) and PPI-120 (SMD = -0.44, 95% CI = [-0.54, -0.33]). The sensitivity and meta-regression analysis showed that sample size, gender proportion, imbalance for gender, source of control group, and study continent were sources of heterogeneity (P < .05) for both PPI-60 and PPI-120 outcomes. Our findings confirm a global sensorimotor gating deficit in schizophrenia patients, with overall moderate effect size for PPI-60 and PPI-120. Methodological consistency should decrease the high level of heterogeneity of PPI results between studies.
Collapse
Affiliation(s)
- Rodrigo San-Martin
- Center for Mathematics, Computation and Cognition, Universidade Federal do ABC, Santo André, Brazil
| | - Leonardo Andrade Castro
- Center for Mathematics, Computation and Cognition, Universidade Federal do ABC, Santo André, Brazil
| | - Paulo Rossi Menezes
- Department of Preventive Medicine, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- Population Mental Health Research Center, Universidade de São Paulo, São Paulo, Brazil
| | - Francisco José Fraga
- Engineering, Modeling and Applied Social Sciences Center, Universidade Federal do ABC, Santo André, Brazil
| | - Priscyla Waleska Simões
- Engineering, Modeling and Applied Social Sciences Center, Universidade Federal do ABC, Santo André, Brazil
| | - Cristiane Salum
- Center for Mathematics, Computation and Cognition, Universidade Federal do ABC, Santo André, Brazil
| |
Collapse
|
6
|
Prepulse Inhibition of the Auditory Startle Reflex Assessment as a Hallmark of Brainstem Sensorimotor Gating Mechanisms. Brain Sci 2020; 10:brainsci10090639. [PMID: 32947873 PMCID: PMC7563436 DOI: 10.3390/brainsci10090639] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/09/2020] [Accepted: 09/14/2020] [Indexed: 12/20/2022] Open
Abstract
When a low-salience stimulus of any type of sensory modality-auditory, visual, tactile-immediately precedes an unexpected startle-like stimulus, such as the acoustic startle reflex, the startle motor reaction becomes less pronounced or is even abolished. This phenomenon is known as prepulse inhibition (PPI), and it provides a quantitative measure of central processing by filtering out irrelevant stimuli. As PPI implies plasticity of a reflex and is related to automatic or attentional processes, depending on the interstimulus intervals, this behavioral paradigm might be considered a potential marker of short- and long-term plasticity. Assessment of PPI is directly related to the examination of neural sensorimotor gating mechanisms, which are plastic-adaptive operations for preventing overstimulation and helping the brain to focus on a specific stimulus among other distracters. Despite their obvious importance in normal brain activity, little is known about the intimate physiology, circuitry, and neurochemistry of sensorimotor gating mechanisms. In this work, we extensively review the current literature focusing on studies that used state-of-the-art techniques to interrogate the neuroanatomy, connectomics, neurotransmitter-receptor functions, and sex-derived differences in the PPI process, and how we can harness it as biological marker in neurological and psychiatric pathology.
Collapse
|
7
|
Khoja S, Asatryan L, Jakowec MW, Davies DL. Dopamine Receptor Blockade Attenuates Purinergic P2X4 Receptor-Mediated Prepulse Inhibition Deficits and Underlying Molecular Mechanisms. Front Cell Neurosci 2019; 13:331. [PMID: 31396053 PMCID: PMC6664007 DOI: 10.3389/fncel.2019.00331] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 07/04/2019] [Indexed: 11/13/2022] Open
Abstract
Sensorimotor gating refers to the ability to filter incoming sensory information in a stimulus-laden environment and disruption of this physiological process has been documented in psychiatric disorders characterized by cognitive aberrations. The effectiveness of current pharmacotherapies for treatment of sensorimotor gating deficits in the patient population still remains controversial. These challenges emphasize the need to better understand the biological underpinnings of sensorimotor gating which could lead to discovery of novel drug targets for therapeutic intervention. Notably, we recently reported a role for purinergic P2X4 receptors (P2X4Rs) in regulation of sensorimotor gating using prepulse inhibition (PPI) of acoustic startle reflex. P2X4Rs are ion channels gated by adenosine-5′-triphosphate (ATP). Ivermectin (IVM) induced PPI deficits in C57BL/6J mice in a P2X4R-specific manner. Furthermore, mice deficient in P2X4Rs [P2X4R knockout (KO)] exhibited PPI deficits that were alleviated by dopamine (DA) receptor antagonists demonstrating an interaction between P2X4Rs and DA receptors in PPI regulation. On the basis of these findings, we hypothesized that increased DA neurotransmission underlies IVM-mediated PPI deficits. To test this hypothesis, we measured the effects of D1 and D2 receptor antagonists, SCH 23390 and raclopride respectively and D1 agonist, SKF 82958 on IVM-mediated PPI deficits. To gain mechanistic insights, we investigated the interaction between IVM and dopaminergic drugs on signaling molecules linked to PPI regulation in the ventral striatum. SCH 23390 significantly attenuated the PPI disruptive effects of IVM to a much greater degree than that of raclopride. SKF 82958 failed to potentiate IVM-mediated PPI disruption. At the molecular level, modulation of D1 receptors altered IVM’s effects on dopamine and cyclic-AMP regulated phosphoprotein of 32 kDa (DARPP-32) phosphorylation. Additionally, IVM interacted with the DA receptors antagonists and SKF 82958 in phosphorylation of Ca2+/calmodulin kinase IIα (CaMKIIα) and its downstream target, neuronal nitric oxide synthase (nNOS). Current findings suggest an involvement for D1 and D2 receptors in IVM-mediated PPI disruption via modulation of DARPP-32, CaMKIIα and nNOS. Taken together, the findings suggest that stimulation of P2X4Rs can lead to DA hyperactivity and disruption of information processing, implicating P2X4Rs as a novel drug target for treatment of psychiatric disorders characterized by sensorimotor gating deficits.
Collapse
Affiliation(s)
- Sheraz Khoja
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Liana Asatryan
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Michael W Jakowec
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Daryl L Davies
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
8
|
Coors A, Brosch M, Kahl E, Khalil R, Michels B, Laub A, Franke K, Gerber B, Fendt M. Rhodiola rosea root extract has antipsychotic-like effects in rodent models of sensorimotor gating. JOURNAL OF ETHNOPHARMACOLOGY 2019; 235:320-328. [PMID: 30776471 DOI: 10.1016/j.jep.2019.02.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 02/14/2019] [Accepted: 02/14/2019] [Indexed: 06/09/2023]
Abstract
UNLABELLED ETHNOPHARMACOLOGICAL RELEVANCE: The plant arctic root (Rhodiola rosea, L.) is growing in northern regions of Europe, Asia and North America. Extracts of R. rosea are used in traditional medicine for various conditions related to nervous system function. According to scientific studies from the last decades, the plant might have potential for use in the treatment of memory impairments, stress and depression, but reports concerning other neuropsychiatric disorders are scarce. AIM OF THE STUDY In this context, our study aimed to examine potential antipsychotic-like effects of R. rosea root extract. MATERIALS AND METHODS We tested the effects of R. rosea root extract on prepulse inhibition in rats and mice. Prepulse inhibition is an established operational measure of sensorimotor gating, which is impaired in schizophrenia and other psychotic disorders. RESULTS R. rosea root extract increased prepulse inhibition in rats and mice. Interestingly, the R. rosea extract had stronger effects in those individual animals that had low baseline levels of prepulse inhibition. Therefore, we performed further experiments in which we pharmacologically induced a prepulse inhibition deficit by two different psychostimulants, either the dopamine D2 receptor agonist apomorphine or the NMDA receptor antagonist dizocilpine (MK-801). Pre-treatment with the R. rosea extract significantly restored both, apomorphine- and dizocilpine-induced prepulse inhibition deficits. CONCLUSIONS The present study demonstrates that R. rosea extract robustly reverses prepulse inhibition deficits in rodents. This suggests antipsychotic-like effects of R. rosea extract. Future studies should focus on the pharmacological mechanisms underlying these effects.
Collapse
Affiliation(s)
- Andreas Coors
- Neuroscience Program, University of Bremen, Germany; Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Germany.
| | - Marcel Brosch
- Integrative Neuroscience Program, Otto-von-Guericke University Magdeburg, Germany.
| | - Evelyn Kahl
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Germany.
| | - Radwa Khalil
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Germany.
| | - Birgit Michels
- Genetics, Leibniz Institute for Neurobiology, Magdeburg, Germany.
| | - Annegret Laub
- Leibniz Institute of Plant Biochemistry, Halle, Germany.
| | - Katrin Franke
- Leibniz Institute of Plant Biochemistry, Halle, Germany.
| | - Bertram Gerber
- Genetics, Leibniz Institute for Neurobiology, Magdeburg, Germany; Institute of Biology, Otto-von-Guericke University Magdeburg, Germany; Center of Behavioral Brain Sciences, Otto-von-Guericke University Magdeburg, Germany.
| | - Markus Fendt
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Germany; Center of Behavioral Brain Sciences, Otto-von-Guericke University Magdeburg, Germany.
| |
Collapse
|
9
|
Prenatal treatment with methylazoxymethanol acetate as a neurodevelopmental disruption model of schizophrenia in mice. Neuropharmacology 2019; 150:1-14. [DOI: 10.1016/j.neuropharm.2019.02.034] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 02/12/2019] [Accepted: 02/25/2019] [Indexed: 12/26/2022]
|
10
|
Abstract
Schizophrenia (SZ) is a severe psychotic disorder that is highly heritable and common in the general population. The genetic heterogeneity of SZ is substantial, with contributions from common, rare, and de novo variants, in addition to environmental factors. Large genome-wide association studies have detected many variants that are associated with SZ, yet the pathways by which these variants influence risk remain largely unknown. SZ is also clinically heterogeneous, with patients exhibiting a broad range of deficits and symptom severity that vary over the course of illness and treatment, which has complicated efforts to identify risk variants. However, the underlying brain dysfunction forms a more stable trait marker that quantitative neurocognitive and neurophysiological endophenotypes may be able to objectively measure. These endophenotypes are less likely to be heterogeneous than the disorder and provide a neurobiological context to detect risk variants and underlying pathways among genes associated with SZ diagnosis. Furthermore, many endophenotypes are translational into animal model systems, allowing for direct evaluation of the neural circuit dysfunctions and neurobiological substrates. We review a selection of the most promising SZ endophenotypes, including prepulse inhibition, mismatch negativity, oculomotor antisaccade, letter-number sequencing, and continuous performance tests. We also highlight recent findings from large consortia that suggest the potential role of genes, particularly in the neuregulin and glutamate pathways, in several of these endophenotypes. Although endophenotypes require additional time and effort to assess, the insight into the underlying neurobiology that they provide may ultimately reveal the underlying genetic architecture for SZ and suggest novel treatment targets.
Collapse
|
11
|
Saletti PG, Tomaz C. Cannabidiol effects on prepulse inhibition in nonhuman primates. Rev Neurosci 2018; 30:95-105. [DOI: 10.1515/revneuro-2017-0101] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 03/29/2018] [Indexed: 01/22/2023]
Abstract
AbstractPrepulse inhibition (PPI) of acoustic startle reflex is a well-established behavior paradigm to measure sensorimotor gating deficits. PPI is disrupted in several neuropsychiatric disorders, including schizophrenia. PPI tests can be used to screen new drugs for treatment of such disorders. In this review, we discuss how PPI paradigm can help in screening the therapeutic effects of cannabidiol (CBD). We look into recent literature about CBD effects on PPI response in animal models, especially in nonhuman primates. CBD has been shown to modify PPI inN-methyld-aspartate receptor antagonist models for schizophrenia, both in rodents and in nonhuman primates. These results show CBD as a potential drug for the treatment of neurologic disorders that present alterations in sensorimotor system, such as schizophrenia. Moreover, the PPI paradigm seems to be a useful and relative simple paradigm to test the efficacy of CBD as a potential therapeutic drug.
Collapse
|
12
|
Fargotstein M, Hasenkamp W, Gross R, Cuthbert B, Green A, Swails L, Lewison B, Boshoven W, Keyes M, Duncan E. The effect of antipsychotic medications on acoustic startle latency in schizophrenia. Schizophr Res 2018; 198:28-35. [PMID: 28732798 DOI: 10.1016/j.schres.2017.07.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 07/06/2017] [Accepted: 07/09/2017] [Indexed: 10/19/2022]
Abstract
Prepulse inhibition of the acoustic startle reflex (PPI) is extensively studied as a biomarker of schizophrenia (SCZ); however, antipsychotic medication can confound the measure. Latency, the time between the startling stimulus and the reflexive eye blink, provides an index of neural processing speed and is 90% heritable. SCZ subjects have slower latency than controls (CON). This study examined the effects of antipsychotic medication on startle latency. 108 CON and 132 SCZ subjects in three medication subgroups (94 on second-generation antipsychotics (SGA), 25 on first-generation antipsychotics (FGA), 13 unmedicated (NoMed)) were tested on a standard acoustic startle paradigm designed to measure startle magnitude, PPI, and latency. Latency was slower in SCZ compared to CON subjects (p=0.005). Latency did not differ between the three SCZ medication groups. When CON were added to that model, both the NoMed subjects (p=0.04) and the SGA subjects (p=0.003) were slower than CON subjects. For PPI, CON did not differ from SCZ analyzed as a single group. When SCZ subjects were divided into medication groups, PPI was lower in NoMed subjects than the CON group (p=0.03), the SGA group (p=0.02) and the FGA group (p=0.05). SCZ subjects on any medication did not differ from CON. Thus, latency was partially normalized by antipsychotic medication, but this did not obscure the slower latency in SCZ compared to CON. Therefore latency is both trait and state related, whereas medication normalized PPI and obscured any difference between SCZ and CON.
Collapse
Affiliation(s)
- Molly Fargotstein
- Atlanta Veterans Affairs Medical Center, 1670 Clairmont Road, Decatur, GA 30033, USA
| | - Wendy Hasenkamp
- Atlanta Veterans Affairs Medical Center, 1670 Clairmont Road, Decatur, GA 30033, USA
| | - Robin Gross
- Atlanta Veterans Affairs Medical Center, 1670 Clairmont Road, Decatur, GA 30033, USA; Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 12 Executive Park Drive Northeast #200, Atlanta, GA 30329, USA
| | - Bruce Cuthbert
- Atlanta Veterans Affairs Medical Center, 1670 Clairmont Road, Decatur, GA 30033, USA; Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 12 Executive Park Drive Northeast #200, Atlanta, GA 30329, USA
| | - Amanda Green
- Atlanta Veterans Affairs Medical Center, 1670 Clairmont Road, Decatur, GA 30033, USA
| | - Lisette Swails
- Atlanta Veterans Affairs Medical Center, 1670 Clairmont Road, Decatur, GA 30033, USA
| | - Barbara Lewison
- Atlanta Veterans Affairs Medical Center, 1670 Clairmont Road, Decatur, GA 30033, USA; Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 12 Executive Park Drive Northeast #200, Atlanta, GA 30329, USA
| | - William Boshoven
- Atlanta Veterans Affairs Medical Center, 1670 Clairmont Road, Decatur, GA 30033, USA; Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 12 Executive Park Drive Northeast #200, Atlanta, GA 30329, USA
| | - Megan Keyes
- Atlanta Veterans Affairs Medical Center, 1670 Clairmont Road, Decatur, GA 30033, USA
| | - Erica Duncan
- Atlanta Veterans Affairs Medical Center, 1670 Clairmont Road, Decatur, GA 30033, USA; Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 12 Executive Park Drive Northeast #200, Atlanta, GA 30329, USA.
| |
Collapse
|
13
|
Swerdlow NR, Light GA. Sensorimotor gating deficits in schizophrenia: Advancing our understanding of the phenotype, its neural circuitry and genetic substrates. Schizophr Res 2018; 198. [PMID: 29525460 PMCID: PMC6103885 DOI: 10.1016/j.schres.2018.02.042] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Neal R Swerdlow
- Department of Psychiatry, University of California, San Diego, School of Medicine, La Jolla, CA, United States.
| | - Gregory A Light
- Department of Psychiatry, University of California, San Diego, School of Medicine, La Jolla, CA, United States; Research Service, VA San Diego Healthcare System, San Diego, CA, United States
| |
Collapse
|
14
|
Sedgwick O, Young S, Greer B, Arnold J, Parsons A, Puzzo I, Terracciano M, Das M, Kumari V. Sensorimotor gating characteristics of violent men with comorbid psychosis and dissocial personality disorder: Relationship with antisocial traits and psychosocial deprivation. Schizophr Res 2018; 198:21-27. [PMID: 28689756 DOI: 10.1016/j.schres.2017.06.045] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 06/20/2017] [Accepted: 06/23/2017] [Indexed: 12/14/2022]
Abstract
Evidence suggests violence amongst those with psychosis is not aetiologically homogeneous, and that a large proportion of those who engage in violent behaviour have a comorbid antisocial personality disorder. Initial investigations indicate that this subgroup has distinct historical and neuropsychological characteristics, which may indicate diverse treatment needs. This study investigated sensorimotor gating characteristics of violent men with diagnoses of both psychosis and dissocial personality disorder (DPD) (n=21) relative to violent men with psychosis alone (n=12), DPD alone (n=14) and healthy, non-violent male controls (n=27), using the prepulse inhibition (PPI) paradigm. The results indicated that, relative to the psychosis alone and healthy control groups, the comorbid group had lower PPI, especially at 60-ms prepulse-to-pulse interval. The DPD group took an intermediary position and did not differ from any group. Antisocial personality traits (factor two scores of the Psychopathy Checklist - Revised), and greater severity of childhood psychosocial deprivation (including physical and sexual abuse), were significantly correlated with poor PPI across the clinical sample. The findings suggest diverse sensorimotor gating profiles amongst subgroups of violent offenders, with comorbid psychosis and DPD showing most impairment. This is consistent with a 'double dose' of deficit explanation amongst those with both diagnoses, explained at least in part by presence of antisocial personality traits and childhood psychosocial deprivation.
Collapse
Affiliation(s)
- Ottilie Sedgwick
- Department of Psychology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK; Forensic Research Domain, Broadmoor Hospital, West London Mental Health Trust, UK.
| | - Susan Young
- Forensic Research Domain, Broadmoor Hospital, West London Mental Health Trust, UK; Centre for Mental Health, Department of Medicine, Imperial College London, UK
| | - Ben Greer
- Forensic Research Domain, Broadmoor Hospital, West London Mental Health Trust, UK
| | - Jack Arnold
- Forensic Research Domain, Broadmoor Hospital, West London Mental Health Trust, UK
| | - Aisling Parsons
- Forensic Research Domain, Broadmoor Hospital, West London Mental Health Trust, UK
| | - Ignazio Puzzo
- Forensic Research Domain, Broadmoor Hospital, West London Mental Health Trust, UK
| | - Mariafatima Terracciano
- Department of Psychology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Mrigendra Das
- Forensic Research Domain, Broadmoor Hospital, West London Mental Health Trust, UK
| | - Veena Kumari
- Division of Psychology, College of Health and Life Sciences, Brunel University London, UK.
| |
Collapse
|
15
|
Swerdlow NR, Light GA, Thomas ML, Sprock J, Calkins ME, Green MF, Greenwood TA, Gur RE, Gur RC, Lazzeroni LC, Nuechterlein KH, Radant AD, Seidman LJ, Siever LJ, Silverman JM, Stone WS, Sugar CA, Tsuang DW, Tsuang MT, Turetsky BI, Braff DL. Deficient prepulse inhibition in schizophrenia in a multi-site cohort: Internal replication and extension. Schizophr Res 2018; 198:6-15. [PMID: 28549722 PMCID: PMC5700873 DOI: 10.1016/j.schres.2017.05.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/08/2017] [Accepted: 05/10/2017] [Indexed: 11/29/2022]
Abstract
BACKGROUND The Consortium on the Genetics of Schizophrenia (COGS) collected case-control endophenotype and genetic information from 2457 patients and healthy subjects (HS) across 5 test sites over 3.5 years. Analysis of the first "wave" (W1) of 1400 subjects identified prepulse inhibition (PPI) deficits in patients vs. HS. Data from the second COGS "wave" (W2), and the combined W(1+2), were used to assess: 1) the replicability of PPI deficits in this design; 2) the impact of response criteria on PPI deficits; and 3) PPI in a large cohort of antipsychotic-free patients. METHODS PPI in W2 HS (n=315) and schizophrenia patients (n=326) was compared to findings from W1; planned analyses assessed the impact of diagnosis, "wave" (1 vs. 2), and startle magnitude criteria. Combining waves allowed us to assess PPI in 120 antipsychotic-free patients, including many in the early course of illness. RESULTS ANOVA of all W(1+2) subjects revealed robust PPI deficits in patients across "waves" (p<0.0004). Strict response criteria excluded almost 39% of all subjects, disproportionately impacting specific subgroups; ANOVA in this smaller cohort confirmed no significant effect of "wave" or "wave x diagnosis" interaction, and a significant effect of diagnosis (p<0.002). Antipsychotic-free, early-illness patients had particularly robust PPI deficits. DISCUSSION Schizophrenia-linked PPI deficits were replicable across two multi-site "waves" of subjects collected over 3.5years. Strict response criteria disproportionately excluded older, male, non-Caucasian patients with low-normal hearing acuity. These findings set the stage for genetic analyses of PPI using the combined COGS wave 1 and 2 cohorts.
Collapse
Affiliation(s)
- Neal R. Swerdlow
- Department of Psychiatry, University of California San Diego, La Jolla, CA,Corresponding Author: Neal R. Swerdlow, M.D., Ph.D., University of California San Diego, Dept. of Psychiatry, 9500 Gilman Drive, La Jolla, CA 92093-0804 619-543-6270 (office); 619-543-2493 (fax);
| | - Gregory A. Light
- Department of Psychiatry, University of California San Diego, La Jolla, CA,VISN 22, Mental Illness Research, Education & Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, CA
| | - Michael L. Thomas
- Department of Psychiatry, University of California San Diego, La Jolla, CA,VISN 22, Mental Illness Research, Education & Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, CA
| | - Joyce Sprock
- Department of Psychiatry, University of California San Diego, La Jolla, CA,VISN 22, Mental Illness Research, Education & Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, CA
| | - Monica E. Calkins
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - Michael F. Green
- Department of Psychiatry and Biobehavioral Sciences, Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA,VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | | | - Raquel E. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - Ruben C. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - Laura C. Lazzeroni
- Departments of Psychiatry and Behavioral Sciences and of Pediatrics, Stanford University, Stanford, CA
| | - Keith H. Nuechterlein
- Department of Psychiatry and Biobehavioral Sciences, Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Allen D. Radant
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA,VA Puget Sound Health Care System, Seattle, WA
| | - Larry J. Seidman
- Department of Psychiatry, Harvard Medical School, Boston, MA,Massachusetts Mental Health Center Public Psychiatry Division of the Beth Israel Deaconess Medical Center, Boston, MA
| | - Larry J. Siever
- Department of Psychiatry, The Mount Sinai School of Medicine, New York, NY,James J. Peters VA Medical Center, New York, NY
| | - Jeremy M. Silverman
- Department of Psychiatry, The Mount Sinai School of Medicine, New York, NY,James J. Peters VA Medical Center, New York, NY
| | - William S. Stone
- Department of Psychiatry, Harvard Medical School, Boston, MA,Harvard Institute of Psychiatric Epidemiology and Genetics, Boston, MA
| | - Catherine A. Sugar
- Department of Psychiatry and Biobehavioral Sciences, Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA,VISN 22, Mental Illness Research, Education & Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, CA,Department of Biostatistics, University of California Los Angeles School of Public Health, Los Angeles, CA
| | - Debby W. Tsuang
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA,VA Puget Sound Health Care System, Seattle, WA
| | - Ming T. Tsuang
- Department of Psychiatry, University of California San Diego, La Jolla, CA,Institute for Genomic Medicine, University of California San Diego, La Jolla, CA,Harvard Institute of Psychiatric Epidemiology and Genetics, Boston, MA
| | - Bruce I. Turetsky
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - David L. Braff
- Department of Psychiatry, University of California San Diego, La Jolla, CA,VISN 22, Mental Illness Research, Education & Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, CA
| |
Collapse
|
16
|
Moszczynski AJ, Gopaul J, McCunn P, Volkening K, Harvey M, Bartha R, Schmid S, Strong MJ. Somatic Gene Transfer Using a Recombinant Adenoviral Vector (rAAV9) Encoding Pseudophosphorylated Human Thr175 Tau in Adult Rat Hippocampus Induces Tau Pathology. J Neuropathol Exp Neurol 2018; 77:685-695. [DOI: 10.1093/jnen/nly044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Alexander J Moszczynski
- Department of Clinical Neurological Sciences, Molecular Medicine Group, Robarts Research Institute
| | | | | | - Kathryn Volkening
- Department of Clinical Neurological Sciences, Molecular Medicine Group, Robarts Research Institute
| | - Madeline Harvey
- Department of Clinical Neurological Sciences, Molecular Medicine Group, Robarts Research Institute
| | | | | | - Michael J Strong
- Department of Clinical Neurological Sciences, Molecular Medicine Group, Robarts Research Institute
- Department of Clinical Neurological Sciences, University Hospital, University of Western Ontario, Ontario, Canada
| |
Collapse
|
17
|
Khan KM, Collier AD, Meshalkina DA, Kysil EV, Khatsko SL, Kolesnikova T, Morzherin YY, Warnick JE, Kalueff AV, Echevarria DJ. Zebrafish models in neuropsychopharmacology and CNS drug discovery. Br J Pharmacol 2017; 174:1925-1944. [PMID: 28217866 PMCID: PMC5466539 DOI: 10.1111/bph.13754] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 02/11/2017] [Accepted: 02/14/2017] [Indexed: 12/12/2022] Open
Abstract
Despite the high prevalence of neuropsychiatric disorders, their aetiology and molecular mechanisms remain poorly understood. The zebrafish (Danio rerio) is increasingly utilized as a powerful animal model in neuropharmacology research and in vivo drug screening. Collectively, this makes zebrafish a useful tool for drug discovery and the identification of disordered molecular pathways. Here, we discuss zebrafish models of selected human neuropsychiatric disorders and drug-induced phenotypes. As well as covering a broad range of brain disorders (from anxiety and psychoses to neurodegeneration), we also summarize recent developments in zebrafish genetics and small molecule screening, which markedly enhance the disease modelling and the discovery of novel drug targets.
Collapse
Affiliation(s)
- Kanza M Khan
- Department of PsychologyUniversity of Southern MississippiHattiesburgMSUSA
| | - Adam D Collier
- Department of PsychologyUniversity of Southern MississippiHattiesburgMSUSA
- The International Zebrafish Neuroscience Research Consortium (ZNRC)SlidellLAUSA
| | - Darya A Meshalkina
- The International Zebrafish Neuroscience Research Consortium (ZNRC)SlidellLAUSA
- Institute of Translational BiomedicineSt. Petersburg State UniversitySt. PetersburgRussia
| | - Elana V Kysil
- Institute of Translational BiomedicineSt. Petersburg State UniversitySt. PetersburgRussia
| | | | | | | | - Jason E Warnick
- The International Zebrafish Neuroscience Research Consortium (ZNRC)SlidellLAUSA
- Department of Behavioral SciencesArkansas Tech UniversityRussellvilleARUSA
| | - Allan V Kalueff
- The International Zebrafish Neuroscience Research Consortium (ZNRC)SlidellLAUSA
- Institute of Translational BiomedicineSt. Petersburg State UniversitySt. PetersburgRussia
- Ural Federal UniversityEkaterinburgRussia
- Research Institute of Marine Drugs and Nutrition, College of Food Science and TechnologyGuangdong Ocean UniversityZhanjiangGuangdongChina
| | - David J Echevarria
- Department of PsychologyUniversity of Southern MississippiHattiesburgMSUSA
- The International Zebrafish Neuroscience Research Consortium (ZNRC)SlidellLAUSA
| |
Collapse
|
18
|
Yang NB, Tian Q, Fan Y, Bo QJ, Zhang L, Li L, Wang CY. Deficits of perceived spatial separation induced prepulse inhibition in patients with schizophrenia: relationships to symptoms and neurocognition. BMC Psychiatry 2017; 17:135. [PMID: 28399842 PMCID: PMC5387250 DOI: 10.1186/s12888-017-1276-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 03/18/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Prepulse inhibition (PPI) and attention were impaired, which may cause psychotic symptoms and (or) hinder the cognitive functions in schizophrenia. However, due to the measurement methods of PPI, findings about the relationship between PPI and clinical symptoms, cognitive performances have been equivocal. METHODS Seventy-five schizophrenia patients (SZ) and 50 healthy controls (HC) were assessed in a modified acoustic PPI paradigm, named perceived spatial separation-induced PPI (PSS-PPI), compared to perceived spatial co-location PPI (PSC-PPI) with inter-stimulus interval (ISI) of 120 ms. Repeatable Battery for the Assessment of Neuropsychological Status and the Stroop Color-Word Test were administered to all subjects. RESULTS Significant decrease in the modified PPI was found in the patients as compared to the controls, and effect sizes (Cohen'd) for patients vs. HCs % PPI levels achieved a significant level (PSC-PPI d = 0.84, PSS-PPI d = 1.27). A logistic regression model based on PSS-PPI significantly represented the diagnostic grouping (χ2= 29.3; p < 0 .001), with 85.2% area under ROC curve in predicting group membership. In addition, patients exhibited deficits in neurocognition. Among patients of "non-remission", after controlling for gender, age, education, duration, recurrence times, onset age, cigarettes per day and chlorpromazine equivalent dosage, PSS-PPI levels were associated with positive and negative symptoms, PANSS total and thought disorder (P1, P6, P7, N5, N7, G9). In multiple linear regression analyses, male and higher attention scores contributed to better PSC-PPI and PSS-PPI in controls group, while larger amount of smoke and longer word-color interfere time contributed to poor PSS-PPI. In patients' group, higher education and attention scores contributed to better PSS-PPI, while repeated relapse contributed to poor PSS-PPI. CONCLUSIONS The acoustic perceived spatial separation-induced PPIs may bring to light the psychopathological symptoms, especially for thought disorder, and the mechanism(s) of the novel PPI paradigm was associated with attention function.
Collapse
Affiliation(s)
- Ning-Bo Yang
- grid.24696.3fDepartment of Psychiatry, Beijing Anding Hospital, Capital Medical University, No.5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088 China ,Beijing Key Laboratory of Mental Disorders, No.5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088 China ,Beijing Institute for Brain Disorders Center of Schizophrenia, No.5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088 China
| | - Qing Tian
- grid.24696.3fDepartment of Psychiatry, Beijing Anding Hospital, Capital Medical University, No.5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088 China ,Beijing Key Laboratory of Mental Disorders, No.5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088 China ,Beijing Institute for Brain Disorders Center of Schizophrenia, No.5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088 China
| | - Yu Fan
- grid.24696.3fDepartment of Psychiatry, Beijing Anding Hospital, Capital Medical University, No.5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088 China ,Beijing Key Laboratory of Mental Disorders, No.5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088 China ,Beijing Institute for Brain Disorders Center of Schizophrenia, No.5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088 China
| | - Qi-Jing Bo
- grid.24696.3fDepartment of Psychiatry, Beijing Anding Hospital, Capital Medical University, No.5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088 China ,Beijing Key Laboratory of Mental Disorders, No.5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088 China ,Beijing Institute for Brain Disorders Center of Schizophrenia, No.5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088 China
| | - Liang Zhang
- grid.24696.3fDepartment of Psychiatry, Beijing Anding Hospital, Capital Medical University, No.5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088 China ,Beijing Key Laboratory of Mental Disorders, No.5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088 China ,Beijing Institute for Brain Disorders Center of Schizophrenia, No.5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088 China
| | - Liang Li
- grid.11135.37Department of Psychology, Peking University, Beijing, 100871 China ,grid.419897.aKey Laboratory on Machine Perception (Ministry of Education), Beijing, 100871 China ,McGovern Institute for Brain Research, Beijing, 100871 China
| | - Chuan-Yue Wang
- Department of Psychiatry, Beijing Anding Hospital, Capital Medical University, No.5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088, China. .,Beijing Key Laboratory of Mental Disorders, No.5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088, China. .,Beijing Institute for Brain Disorders Center of Schizophrenia, No.5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088, China.
| |
Collapse
|
19
|
Ronca SE, Smith J, Koma T, Miller MM, Yun N, Dineley KT, Paessler S. Mouse Model of Neurological Complications Resulting from Encephalitic Alphavirus Infection. Front Microbiol 2017; 8:188. [PMID: 28223982 PMCID: PMC5293790 DOI: 10.3389/fmicb.2017.00188] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 01/25/2017] [Indexed: 01/20/2023] Open
Abstract
Long-term neurological complications, termed sequelae, can result from viral encephalitis, which are not well understood. In human survivors, alphavirus encephalitis can cause severe neurobehavioral changes, in the most extreme cases, a schizophrenic-like syndrome. In the present study, we aimed to adapt an animal model of alphavirus infection survival to study the development of these long-term neurological complications. Upon low-dose infection of wild-type C57B/6 mice, asymptomatic and symptomatic groups were established and compared to mock-infected mice to measure general health and baseline neurological function, including the acoustic startle response and prepulse inhibition paradigm. Prepulse inhibition is a robust operational measure of sensorimotor gating, a fundamental form of information processing. Deficits in prepulse inhibition manifest as the inability to filter out extraneous sensory stimuli. Sensory gating is disrupted in schizophrenia and other mental disorders, as well as neurodegenerative diseases. Symptomatic mice developed deficits in prepulse inhibition that lasted through 6 months post infection; these deficits were absent in asymptomatic or mock-infected groups. Accompanying prepulse inhibition deficits, symptomatic animals exhibited thalamus damage as visualized with H&E staining, as well as increased GFAP expression in the posterior complex of the thalamus and dentate gyrus of the hippocampus. These histological changes and increased GFAP expression were absent in the asymptomatic and mock-infected animals, indicating that glial scarring could have contributed to the prepulse inhibition phenotype observed in the symptomatic animals. This model provides a tool to test mechanisms of and treatments for the neurological sequelae of viral encephalitis and begins to delineate potential explanations for the development of such sequelae post infection.
Collapse
Affiliation(s)
- Shannon E Ronca
- Department of Pathology, University of Texas Medical Branch, GalvestonTX, USA; Department of Preventive Medicine and Community Health, University of Texas Medical Branch, GalvestonTX, USA
| | - Jeanon Smith
- Department of Pathology, University of Texas Medical Branch, Galveston TX, USA
| | - Takaaki Koma
- Department of Pathology, University of Texas Medical Branch, Galveston TX, USA
| | - Magda M Miller
- Department of Pathology, University of Texas Medical Branch, Galveston TX, USA
| | - Nadezhda Yun
- Department of Pathology, University of Texas Medical Branch, Galveston TX, USA
| | - Kelly T Dineley
- Department of Neurology, Center for Addiction Research, University of Texas Medical Branch, Galveston TX, USA
| | - Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch, GalvestonTX, USA; Galveston National Lab, Institute for Human Infections and Immunity, University of Texas Medical Branch, GalvestonTX, USA
| |
Collapse
|
20
|
Didriksen M, Fejgin K, Nilsson SR, Birknow MR, Grayton HM, Larsen PH, Lauridsen JB, Nielsen V, Celada P, Santana N, Kallunki P, Christensen KV, Werge TM, Stensbøl TB, Egebjerg J, Gastambide F, Artigas F, Bastlund JF, Nielsen J. Persistent gating deficit and increased sensitivity to NMDA receptor antagonism after puberty in a new mouse model of the human 22q11.2 microdeletion syndrome: a study in male mice. J Psychiatry Neurosci 2017; 42:48-58. [PMID: 27391101 PMCID: PMC5373712 DOI: 10.1503/jpn.150381] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 03/05/2016] [Accepted: 04/05/2016] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND The hemizygous 22q11.2 microdeletion is a common copy number variant in humans. The deletion confers high risk for neurodevelopmental disorders, including autism and schizophrenia. Up to 41% of deletion carriers experience psychotic symptoms. METHODS We present a new mouse model (Df(h22q11)/+) of the deletion syndrome (22q11.2DS) and report on, to our knowledge, the most comprehensive study undertaken to date in 22q11.2DS models. The study was conducted in male mice. RESULTS We found elevated postpubertal N-methyl-D-aspartate (NMDA) receptor antagonist-induced hyperlocomotion, age-independent prepulse inhibition (PPI) deficits and increased acoustic startle response (ASR). The PPI deficit and increased ASR were resistant to antipsychotic treatment. The PPI deficit was not a consequence of impaired hearing measured by auditory brain stem responses. The Df(h22q11)/+ mice also displayed increased amplitude of loudness-dependent auditory evoked potentials. Prefrontal cortex and dorsal striatal elevations of the dopamine metabolite DOPAC and increased dorsal striatal expression of the AMPA receptor subunit GluR1 was found. The Df(h22q11)/+ mice did not deviate from wild-type mice in a wide range of other behavioural and biochemical assays. LIMITATIONS The 22q11.2 microdeletion has incomplete penetrance in humans, and the severity of disease depends on the complete genetic makeup in concert with environmental factors. In order to obtain more marked phenotypes reflecting the severe conditions related to 22q11.2DS it is suggested to expose the Df(h22q11)/+ mice to environmental stressors that may unmask latent psychopathology. CONCLUSION The Df(h22q11)/+ model will be a valuable tool for increasing our understanding of the etiology of schizophrenia and other psychiatric disorders associated with the 22q11DS.
Collapse
Affiliation(s)
- Michael Didriksen
- From H. Lundbeck A/S, Research DK, Valby, Denmark (Didriksen Fejgin, Birknow, Larsen, Lauridsen, Nielsen, Kallunki, Christensen, Stensbøl, Egebjerg, Nielsen); the Department of Psychology, University of Cambridge, Cambridge, UK (Nilsson); the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK (Nilsson); the Lilly Centre for Cognitive Neuroscience, Lilly Research Laboratories, Windlesham, UK (Grayton, Gastambide); the Institut d’Investigacions Biomèdiques de Barcelona, Barcelona, Spain (Celada, Artigas); the Centro de Investigación Biomédica en Red de Salud Mental, Spain (Santana, Artigas); the Institute of Biological Psychiatry, MHC Sct. Hans, Copenhagen Mental Health Services; and the Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen; iP-SYCH - The Lundbeck Foundation’s Initiative for Integrative Psychiatric Research, Roskilde, Denmark (Werge)
| | - Kim Fejgin
- From H. Lundbeck A/S, Research DK, Valby, Denmark (Didriksen Fejgin, Birknow, Larsen, Lauridsen, Nielsen, Kallunki, Christensen, Stensbøl, Egebjerg, Nielsen); the Department of Psychology, University of Cambridge, Cambridge, UK (Nilsson); the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK (Nilsson); the Lilly Centre for Cognitive Neuroscience, Lilly Research Laboratories, Windlesham, UK (Grayton, Gastambide); the Institut d’Investigacions Biomèdiques de Barcelona, Barcelona, Spain (Celada, Artigas); the Centro de Investigación Biomédica en Red de Salud Mental, Spain (Santana, Artigas); the Institute of Biological Psychiatry, MHC Sct. Hans, Copenhagen Mental Health Services; and the Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen; iP-SYCH - The Lundbeck Foundation’s Initiative for Integrative Psychiatric Research, Roskilde, Denmark (Werge)
| | - Simon R.O. Nilsson
- From H. Lundbeck A/S, Research DK, Valby, Denmark (Didriksen Fejgin, Birknow, Larsen, Lauridsen, Nielsen, Kallunki, Christensen, Stensbøl, Egebjerg, Nielsen); the Department of Psychology, University of Cambridge, Cambridge, UK (Nilsson); the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK (Nilsson); the Lilly Centre for Cognitive Neuroscience, Lilly Research Laboratories, Windlesham, UK (Grayton, Gastambide); the Institut d’Investigacions Biomèdiques de Barcelona, Barcelona, Spain (Celada, Artigas); the Centro de Investigación Biomédica en Red de Salud Mental, Spain (Santana, Artigas); the Institute of Biological Psychiatry, MHC Sct. Hans, Copenhagen Mental Health Services; and the Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen; iP-SYCH - The Lundbeck Foundation’s Initiative for Integrative Psychiatric Research, Roskilde, Denmark (Werge)
| | - Michelle R. Birknow
- From H. Lundbeck A/S, Research DK, Valby, Denmark (Didriksen Fejgin, Birknow, Larsen, Lauridsen, Nielsen, Kallunki, Christensen, Stensbøl, Egebjerg, Nielsen); the Department of Psychology, University of Cambridge, Cambridge, UK (Nilsson); the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK (Nilsson); the Lilly Centre for Cognitive Neuroscience, Lilly Research Laboratories, Windlesham, UK (Grayton, Gastambide); the Institut d’Investigacions Biomèdiques de Barcelona, Barcelona, Spain (Celada, Artigas); the Centro de Investigación Biomédica en Red de Salud Mental, Spain (Santana, Artigas); the Institute of Biological Psychiatry, MHC Sct. Hans, Copenhagen Mental Health Services; and the Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen; iP-SYCH - The Lundbeck Foundation’s Initiative for Integrative Psychiatric Research, Roskilde, Denmark (Werge)
| | - Hannah M. Grayton
- From H. Lundbeck A/S, Research DK, Valby, Denmark (Didriksen Fejgin, Birknow, Larsen, Lauridsen, Nielsen, Kallunki, Christensen, Stensbøl, Egebjerg, Nielsen); the Department of Psychology, University of Cambridge, Cambridge, UK (Nilsson); the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK (Nilsson); the Lilly Centre for Cognitive Neuroscience, Lilly Research Laboratories, Windlesham, UK (Grayton, Gastambide); the Institut d’Investigacions Biomèdiques de Barcelona, Barcelona, Spain (Celada, Artigas); the Centro de Investigación Biomédica en Red de Salud Mental, Spain (Santana, Artigas); the Institute of Biological Psychiatry, MHC Sct. Hans, Copenhagen Mental Health Services; and the Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen; iP-SYCH - The Lundbeck Foundation’s Initiative for Integrative Psychiatric Research, Roskilde, Denmark (Werge)
| | - Peter H. Larsen
- From H. Lundbeck A/S, Research DK, Valby, Denmark (Didriksen Fejgin, Birknow, Larsen, Lauridsen, Nielsen, Kallunki, Christensen, Stensbøl, Egebjerg, Nielsen); the Department of Psychology, University of Cambridge, Cambridge, UK (Nilsson); the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK (Nilsson); the Lilly Centre for Cognitive Neuroscience, Lilly Research Laboratories, Windlesham, UK (Grayton, Gastambide); the Institut d’Investigacions Biomèdiques de Barcelona, Barcelona, Spain (Celada, Artigas); the Centro de Investigación Biomédica en Red de Salud Mental, Spain (Santana, Artigas); the Institute of Biological Psychiatry, MHC Sct. Hans, Copenhagen Mental Health Services; and the Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen; iP-SYCH - The Lundbeck Foundation’s Initiative for Integrative Psychiatric Research, Roskilde, Denmark (Werge)
| | - Jes B. Lauridsen
- From H. Lundbeck A/S, Research DK, Valby, Denmark (Didriksen Fejgin, Birknow, Larsen, Lauridsen, Nielsen, Kallunki, Christensen, Stensbøl, Egebjerg, Nielsen); the Department of Psychology, University of Cambridge, Cambridge, UK (Nilsson); the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK (Nilsson); the Lilly Centre for Cognitive Neuroscience, Lilly Research Laboratories, Windlesham, UK (Grayton, Gastambide); the Institut d’Investigacions Biomèdiques de Barcelona, Barcelona, Spain (Celada, Artigas); the Centro de Investigación Biomédica en Red de Salud Mental, Spain (Santana, Artigas); the Institute of Biological Psychiatry, MHC Sct. Hans, Copenhagen Mental Health Services; and the Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen; iP-SYCH - The Lundbeck Foundation’s Initiative for Integrative Psychiatric Research, Roskilde, Denmark (Werge)
| | - Vibeke Nielsen
- From H. Lundbeck A/S, Research DK, Valby, Denmark (Didriksen Fejgin, Birknow, Larsen, Lauridsen, Nielsen, Kallunki, Christensen, Stensbøl, Egebjerg, Nielsen); the Department of Psychology, University of Cambridge, Cambridge, UK (Nilsson); the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK (Nilsson); the Lilly Centre for Cognitive Neuroscience, Lilly Research Laboratories, Windlesham, UK (Grayton, Gastambide); the Institut d’Investigacions Biomèdiques de Barcelona, Barcelona, Spain (Celada, Artigas); the Centro de Investigación Biomédica en Red de Salud Mental, Spain (Santana, Artigas); the Institute of Biological Psychiatry, MHC Sct. Hans, Copenhagen Mental Health Services; and the Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen; iP-SYCH - The Lundbeck Foundation’s Initiative for Integrative Psychiatric Research, Roskilde, Denmark (Werge)
| | - Pau Celada
- From H. Lundbeck A/S, Research DK, Valby, Denmark (Didriksen Fejgin, Birknow, Larsen, Lauridsen, Nielsen, Kallunki, Christensen, Stensbøl, Egebjerg, Nielsen); the Department of Psychology, University of Cambridge, Cambridge, UK (Nilsson); the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK (Nilsson); the Lilly Centre for Cognitive Neuroscience, Lilly Research Laboratories, Windlesham, UK (Grayton, Gastambide); the Institut d’Investigacions Biomèdiques de Barcelona, Barcelona, Spain (Celada, Artigas); the Centro de Investigación Biomédica en Red de Salud Mental, Spain (Santana, Artigas); the Institute of Biological Psychiatry, MHC Sct. Hans, Copenhagen Mental Health Services; and the Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen; iP-SYCH - The Lundbeck Foundation’s Initiative for Integrative Psychiatric Research, Roskilde, Denmark (Werge)
| | - Noemi Santana
- From H. Lundbeck A/S, Research DK, Valby, Denmark (Didriksen Fejgin, Birknow, Larsen, Lauridsen, Nielsen, Kallunki, Christensen, Stensbøl, Egebjerg, Nielsen); the Department of Psychology, University of Cambridge, Cambridge, UK (Nilsson); the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK (Nilsson); the Lilly Centre for Cognitive Neuroscience, Lilly Research Laboratories, Windlesham, UK (Grayton, Gastambide); the Institut d’Investigacions Biomèdiques de Barcelona, Barcelona, Spain (Celada, Artigas); the Centro de Investigación Biomédica en Red de Salud Mental, Spain (Santana, Artigas); the Institute of Biological Psychiatry, MHC Sct. Hans, Copenhagen Mental Health Services; and the Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen; iP-SYCH - The Lundbeck Foundation’s Initiative for Integrative Psychiatric Research, Roskilde, Denmark (Werge)
| | - Pekka Kallunki
- From H. Lundbeck A/S, Research DK, Valby, Denmark (Didriksen Fejgin, Birknow, Larsen, Lauridsen, Nielsen, Kallunki, Christensen, Stensbøl, Egebjerg, Nielsen); the Department of Psychology, University of Cambridge, Cambridge, UK (Nilsson); the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK (Nilsson); the Lilly Centre for Cognitive Neuroscience, Lilly Research Laboratories, Windlesham, UK (Grayton, Gastambide); the Institut d’Investigacions Biomèdiques de Barcelona, Barcelona, Spain (Celada, Artigas); the Centro de Investigación Biomédica en Red de Salud Mental, Spain (Santana, Artigas); the Institute of Biological Psychiatry, MHC Sct. Hans, Copenhagen Mental Health Services; and the Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen; iP-SYCH - The Lundbeck Foundation’s Initiative for Integrative Psychiatric Research, Roskilde, Denmark (Werge)
| | - Kenneth V. Christensen
- From H. Lundbeck A/S, Research DK, Valby, Denmark (Didriksen Fejgin, Birknow, Larsen, Lauridsen, Nielsen, Kallunki, Christensen, Stensbøl, Egebjerg, Nielsen); the Department of Psychology, University of Cambridge, Cambridge, UK (Nilsson); the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK (Nilsson); the Lilly Centre for Cognitive Neuroscience, Lilly Research Laboratories, Windlesham, UK (Grayton, Gastambide); the Institut d’Investigacions Biomèdiques de Barcelona, Barcelona, Spain (Celada, Artigas); the Centro de Investigación Biomédica en Red de Salud Mental, Spain (Santana, Artigas); the Institute of Biological Psychiatry, MHC Sct. Hans, Copenhagen Mental Health Services; and the Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen; iP-SYCH - The Lundbeck Foundation’s Initiative for Integrative Psychiatric Research, Roskilde, Denmark (Werge)
| | - Thomas M. Werge
- From H. Lundbeck A/S, Research DK, Valby, Denmark (Didriksen Fejgin, Birknow, Larsen, Lauridsen, Nielsen, Kallunki, Christensen, Stensbøl, Egebjerg, Nielsen); the Department of Psychology, University of Cambridge, Cambridge, UK (Nilsson); the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK (Nilsson); the Lilly Centre for Cognitive Neuroscience, Lilly Research Laboratories, Windlesham, UK (Grayton, Gastambide); the Institut d’Investigacions Biomèdiques de Barcelona, Barcelona, Spain (Celada, Artigas); the Centro de Investigación Biomédica en Red de Salud Mental, Spain (Santana, Artigas); the Institute of Biological Psychiatry, MHC Sct. Hans, Copenhagen Mental Health Services; and the Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen; iP-SYCH - The Lundbeck Foundation’s Initiative for Integrative Psychiatric Research, Roskilde, Denmark (Werge)
| | - Tine B. Stensbøl
- From H. Lundbeck A/S, Research DK, Valby, Denmark (Didriksen Fejgin, Birknow, Larsen, Lauridsen, Nielsen, Kallunki, Christensen, Stensbøl, Egebjerg, Nielsen); the Department of Psychology, University of Cambridge, Cambridge, UK (Nilsson); the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK (Nilsson); the Lilly Centre for Cognitive Neuroscience, Lilly Research Laboratories, Windlesham, UK (Grayton, Gastambide); the Institut d’Investigacions Biomèdiques de Barcelona, Barcelona, Spain (Celada, Artigas); the Centro de Investigación Biomédica en Red de Salud Mental, Spain (Santana, Artigas); the Institute of Biological Psychiatry, MHC Sct. Hans, Copenhagen Mental Health Services; and the Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen; iP-SYCH - The Lundbeck Foundation’s Initiative for Integrative Psychiatric Research, Roskilde, Denmark (Werge)
| | - Jan Egebjerg
- From H. Lundbeck A/S, Research DK, Valby, Denmark (Didriksen Fejgin, Birknow, Larsen, Lauridsen, Nielsen, Kallunki, Christensen, Stensbøl, Egebjerg, Nielsen); the Department of Psychology, University of Cambridge, Cambridge, UK (Nilsson); the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK (Nilsson); the Lilly Centre for Cognitive Neuroscience, Lilly Research Laboratories, Windlesham, UK (Grayton, Gastambide); the Institut d’Investigacions Biomèdiques de Barcelona, Barcelona, Spain (Celada, Artigas); the Centro de Investigación Biomédica en Red de Salud Mental, Spain (Santana, Artigas); the Institute of Biological Psychiatry, MHC Sct. Hans, Copenhagen Mental Health Services; and the Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen; iP-SYCH - The Lundbeck Foundation’s Initiative for Integrative Psychiatric Research, Roskilde, Denmark (Werge)
| | - Francois Gastambide
- From H. Lundbeck A/S, Research DK, Valby, Denmark (Didriksen Fejgin, Birknow, Larsen, Lauridsen, Nielsen, Kallunki, Christensen, Stensbøl, Egebjerg, Nielsen); the Department of Psychology, University of Cambridge, Cambridge, UK (Nilsson); the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK (Nilsson); the Lilly Centre for Cognitive Neuroscience, Lilly Research Laboratories, Windlesham, UK (Grayton, Gastambide); the Institut d’Investigacions Biomèdiques de Barcelona, Barcelona, Spain (Celada, Artigas); the Centro de Investigación Biomédica en Red de Salud Mental, Spain (Santana, Artigas); the Institute of Biological Psychiatry, MHC Sct. Hans, Copenhagen Mental Health Services; and the Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen; iP-SYCH - The Lundbeck Foundation’s Initiative for Integrative Psychiatric Research, Roskilde, Denmark (Werge)
| | - Francesc Artigas
- From H. Lundbeck A/S, Research DK, Valby, Denmark (Didriksen Fejgin, Birknow, Larsen, Lauridsen, Nielsen, Kallunki, Christensen, Stensbøl, Egebjerg, Nielsen); the Department of Psychology, University of Cambridge, Cambridge, UK (Nilsson); the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK (Nilsson); the Lilly Centre for Cognitive Neuroscience, Lilly Research Laboratories, Windlesham, UK (Grayton, Gastambide); the Institut d’Investigacions Biomèdiques de Barcelona, Barcelona, Spain (Celada, Artigas); the Centro de Investigación Biomédica en Red de Salud Mental, Spain (Santana, Artigas); the Institute of Biological Psychiatry, MHC Sct. Hans, Copenhagen Mental Health Services; and the Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen; iP-SYCH - The Lundbeck Foundation’s Initiative for Integrative Psychiatric Research, Roskilde, Denmark (Werge)
| | - Jesper F. Bastlund
- From H. Lundbeck A/S, Research DK, Valby, Denmark (Didriksen Fejgin, Birknow, Larsen, Lauridsen, Nielsen, Kallunki, Christensen, Stensbøl, Egebjerg, Nielsen); the Department of Psychology, University of Cambridge, Cambridge, UK (Nilsson); the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK (Nilsson); the Lilly Centre for Cognitive Neuroscience, Lilly Research Laboratories, Windlesham, UK (Grayton, Gastambide); the Institut d’Investigacions Biomèdiques de Barcelona, Barcelona, Spain (Celada, Artigas); the Centro de Investigación Biomédica en Red de Salud Mental, Spain (Santana, Artigas); the Institute of Biological Psychiatry, MHC Sct. Hans, Copenhagen Mental Health Services; and the Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen; iP-SYCH - The Lundbeck Foundation’s Initiative for Integrative Psychiatric Research, Roskilde, Denmark (Werge)
| | - Jacob Nielsen
- From H. Lundbeck A/S, Research DK, Valby, Denmark (Didriksen Fejgin, Birknow, Larsen, Lauridsen, Nielsen, Kallunki, Christensen, Stensbøl, Egebjerg, Nielsen); the Department of Psychology, University of Cambridge, Cambridge, UK (Nilsson); the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK (Nilsson); the Lilly Centre for Cognitive Neuroscience, Lilly Research Laboratories, Windlesham, UK (Grayton, Gastambide); the Institut d’Investigacions Biomèdiques de Barcelona, Barcelona, Spain (Celada, Artigas); the Centro de Investigación Biomédica en Red de Salud Mental, Spain (Santana, Artigas); the Institute of Biological Psychiatry, MHC Sct. Hans, Copenhagen Mental Health Services; and the Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen; iP-SYCH - The Lundbeck Foundation’s Initiative for Integrative Psychiatric Research, Roskilde, Denmark (Werge)
| |
Collapse
|
21
|
Swerdlow NR, Braff DL, Geyer MA. Sensorimotor gating of the startle reflex: what we said 25 years ago, what has happened since then, and what comes next. J Psychopharmacol 2016; 30:1072-1081. [PMID: 27539931 PMCID: PMC6036900 DOI: 10.1177/0269881116661075] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Our 1992 paper, 'The neural substrates of sensorimotor gating of the startle reflex: a review of recent findings and their implications', reviewed a series of (then) new and preliminary findings from cross-species studies of prepulse inhibition of the startle reflex, and commented on their implications. At the time that the report was composed, PubMed listed about 40 citations for studies using the search term 'prepulse inhibition'. In the ensuing 25 years, the field has added about 2700 such reports, reflecting the substantial growth in interest in prepulse inhibition and its utility across a number of different experimental applications. The 30th anniversary of the Journal of Psychopharmacology provides an opportunity to comment briefly on what was described in that 1992 report, how the field has progressed in the subsequent decades, and the paths forward for studies of prepulse inhibition and its use as an operational measure of sensorimotor gating. Among these future paths, we highlight the use of prepulse inhibition as: an endophenotype for genomic studies, and a biomarker for healthy brain circuitry, which may predict sensitivity to psychotherapeutics. Our 1992 report was highly speculative and based on paper-thin empirical data, yet viewed in a certain light, it appears to have contained a basic roadmap for a journey spanning the next 25 years of prepulse inhibition research… and 'what a long, strange trip it's been'.
Collapse
Affiliation(s)
- Neal R Swerdlow
- Department of Psychiatry, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - David L Braff
- Department of Psychiatry, University of California San Diego School of Medicine, La Jolla, CA, USA
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Mark A Geyer
- Department of Psychiatry, University of California San Diego School of Medicine, La Jolla, CA, USA
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
22
|
Mena A, Ruiz-Salas JC, Puentes A, Dorado I, Ruiz-Veguilla M, De la Casa LG. Reduced Prepulse Inhibition as a Biomarker of Schizophrenia. Front Behav Neurosci 2016; 10:202. [PMID: 27803654 PMCID: PMC5067522 DOI: 10.3389/fnbeh.2016.00202] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 10/04/2016] [Indexed: 01/24/2023] Open
Abstract
The startle response is composed by a set of reflex behaviors intended to prepare the organism to face a potentially relevant stimulus. This response can be modulated by several factors as, for example, repeated presentations of the stimulus (startle habituation), or by previous presentation of a weak stimulus (Prepulse Inhibition [PPI]). Both phenomena appear disrupted in schizophrenia that is thought to reflect an alteration in dopaminergic and glutamatergic neurotransmission. In this paper we analyze whether the reported deficits are indicating a transient effect restricted to the acute phase of the disease, or if it reflects a more general biomarker or endophenotype of the disorder. To this end, we measured startle responses in the same set of thirteen schizophrenia patients with a cross-sectional design at two periods: 5 days after hospital admission and 3 months after discharge. The results showed that both startle habituation and PPI were impaired in the schizophrenia patients at the acute stage as compared to a control group composed by 13 healthy participants, and that PPI but not startle habituation remained disrupted when registered 3 months after the discharge. These data point to the consideration of PPI, but not startle habituation, as a schizophrenia biomarker.
Collapse
Affiliation(s)
- Auxiliadora Mena
- Department of Experimental Psychology, University of Seville Seville, Spain
| | - Juan C Ruiz-Salas
- Department of Experimental Psychology, University of Seville Seville, Spain
| | - Andrea Puentes
- Neurosciences Institute, El Bosque University Bogotá, Colombia
| | - Inmaculada Dorado
- Institute of Biomedicine, University Hospital Virgen del Rocío Seville, Spain
| | | | - Luis G De la Casa
- Department of Experimental Psychology, University of Seville Seville, Spain
| |
Collapse
|
23
|
Peres FF, Levin R, Almeida V, Zuardi AW, Hallak JE, Crippa JA, Abilio VC. Cannabidiol, among Other Cannabinoid Drugs, Modulates Prepulse Inhibition of Startle in the SHR Animal Model: Implications for Schizophrenia Pharmacotherapy. Front Pharmacol 2016; 7:303. [PMID: 27667973 PMCID: PMC5016523 DOI: 10.3389/fphar.2016.00303] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/26/2016] [Indexed: 02/06/2023] Open
Abstract
Schizophrenia is a severe psychiatric disorder that involves positive, negative and cognitive symptoms. Prepulse inhibition of startle reflex (PPI) is a paradigm that assesses the sensorimotor gating functioning and is impaired in schizophrenia patients as well as in animal models of this disorder. Recent data point to the participation of the endocannabinoid system in the pathophysiology and pharmacotherapy of schizophrenia. Here, we focus on the effects of cannabinoid drugs on the PPI deficit of animal models of schizophrenia, with greater focus on the SHR (Spontaneously Hypertensive Rats) strain, and on the future prospects resulting from these findings.
Collapse
Affiliation(s)
- Fernanda F Peres
- Interdisciplinary Laboratory of Clinical Neurosciences, Department of Psychiatry, Escola Paulista De Medicina, Federal University of São PauloSão Paulo, Brazil; Department of Pharmacology, Escola Paulista De Medicina, Federal University of São PauloSão Paulo, Brazil
| | - Raquel Levin
- Interdisciplinary Laboratory of Clinical Neurosciences, Department of Psychiatry, Escola Paulista De Medicina, Federal University of São PauloSão Paulo, Brazil; Department of Pharmacology, Escola Paulista De Medicina, Federal University of São PauloSão Paulo, Brazil
| | - Valéria Almeida
- Interdisciplinary Laboratory of Clinical Neurosciences, Department of Psychiatry, Escola Paulista De Medicina, Federal University of São PauloSão Paulo, Brazil; Department of Pharmacology, Escola Paulista De Medicina, Federal University of São PauloSão Paulo, Brazil
| | - Antonio W Zuardi
- Department of Neuroscience and Behavior, University of São PauloRibeirão Preto, Brazil; National Institute for Translational Medicine (INCT-TM, CNPq)Ribeirão Preto, Brazil
| | - Jaime E Hallak
- Department of Neuroscience and Behavior, University of São PauloRibeirão Preto, Brazil; National Institute for Translational Medicine (INCT-TM, CNPq)Ribeirão Preto, Brazil
| | - José A Crippa
- Department of Neuroscience and Behavior, University of São PauloRibeirão Preto, Brazil; National Institute for Translational Medicine (INCT-TM, CNPq)Ribeirão Preto, Brazil
| | - Vanessa C Abilio
- Interdisciplinary Laboratory of Clinical Neurosciences, Department of Psychiatry, Escola Paulista De Medicina, Federal University of São PauloSão Paulo, Brazil; Department of Pharmacology, Escola Paulista De Medicina, Federal University of São PauloSão Paulo, Brazil
| |
Collapse
|
24
|
Electroconvulsive shock attenuated microgliosis and astrogliosis in the hippocampus and ameliorated schizophrenia-like behavior of Gunn rat. J Neuroinflammation 2016; 13:230. [PMID: 27590010 PMCID: PMC5009533 DOI: 10.1186/s12974-016-0688-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 08/18/2016] [Indexed: 11/23/2022] Open
Abstract
Background Although electroconvulsive therapy (ECT) is regarded as one of the efficient treatments for intractable psychiatric disorders, the mechanism of therapeutic action remains unclear. Recently, many studies indicate that ECT affects the immune-related cells, such as microglia, astrocytes, and lymphocytes. Moreover, microglial activation and astrocytic activation have been implicated in the postmortem brains of schizophrenia patients. We previously demonstrated that Gunn rats showed schizophrenia-like behavior and microglial activation in their brains. The present study examined the effects of electroconvulsive shock (ECS), an animal counterpart of ECT, on schizophrenia-like behavior, microgliosis, and astrogliosis in the brain of Gunn rats. Methods The rats were divided into four groups, i.e., Wistar sham, Wistar ECS, Gunn sham, and Gunn ECS. ECS groups received ECS once daily for six consecutive days. Subsequently, prepulse inhibition (PPI) test was performed, and immunohistochemistry analysis was carried out to determine the activation degree of microglia and astrocytes in the hippocampus by using anti-CD11b and anti-glial fibrillary acidic protein (GFAP) antibody, respectively. Results We found PPI deficit in Gunn rats compared to Wistar rats, and it was significantly improved by ECS. Immunohistochemistry analysis revealed that immunoreactivity of CD11b and GFAP was significantly increased in Gunn rats compared to Wistar rats. ECS significantly attenuated the immunoreactivity of both CD11b and GFAP in Gunn rats. Conclusions ECS ameliorated schizophrenia-like behavior of Gunn rats and attenuated microgliosis and astrogliosis in the hippocampus of Gunn rats. Accordingly, therapeutic effects of ECT may be exerted, at least in part, by inhibition of glial activation. These results may provide crucial information to elucidate the role of activated glia in the pathogenesis of schizophrenia and to determine whether future therapeutic interventions should attempt to up-regulate or down-regulate glial functions. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0688-2) contains supplementary material, which is available to authorized users.
Collapse
|
25
|
Ronca SE, Dineley KT, Paessler S. Neurological Sequelae Resulting from Encephalitic Alphavirus Infection. Front Microbiol 2016; 7:959. [PMID: 27379085 PMCID: PMC4913092 DOI: 10.3389/fmicb.2016.00959] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/02/2016] [Indexed: 12/17/2022] Open
Abstract
The recent surge in viral clinical cases and associated neurological deficits have reminded us that viral infections can lead to detrimental, long-term effects, termed sequelae, in survivors. Alphaviruses are enveloped, single-stranded positive-sense RNA viruses in the Togaviridae family. Transmission of alphaviruses between and within species occurs mainly via the bite of an infected mosquito bite, giving alphaviruses a place among arboviruses, or arthropod-borne viruses. Alphaviruses are found throughout the world and typically cause arthralgic or encephalitic disease in infected humans. Originally detected in the 1930s, today the major encephalitic viruses include Venezuelan, Western, and Eastern equine encephalitis viruses (VEEV, WEEV, and EEEV, respectively). VEEV, WEEV, and EEEV are endemic to the Americas and are important human pathogens, leading to thousands of human infections each year. Despite awareness of these viruses for nearly 100 years, we possess little mechanistic understanding regarding the complications (sequelae) that emerge after resolution of acute infection. Neurological sequelae are those complications involving damage to the central nervous system that results in cognitive, sensory, or motor deficits that may also manifest as emotional instability and seizures in the most severe cases. This article serves to provide an overview of clinical cases documented in the past century as well as a summary of the reported neurological sequelae due to VEEV, WEEV, and EEEV infection. We conclude with a treatise on the utility of, and practical considerations for animal models applied to the problem of neurological sequelae of viral encephalopathies in order to decipher mechanisms and interventional strategies.
Collapse
Affiliation(s)
- Shannon E Ronca
- Department of Pathology, University of Texas Medical Branch, Galveston, TXUSA; Department of Preventive Medicine and Community Health, University of Texas Medical Branch, Galveston, TXUSA
| | - Kelly T Dineley
- Department of Neurology, Center for Addiction Research, Rodent In Vivo Assessment Core, Mitchell Center for Neurodegenerative Disorders, University of Texas Medical Branch, Galveston, TX USA
| | - Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch, Galveston, TXUSA; Institute for Human Infections and Immunity, Galveston National Laboratory, University of Texas Medical Branch, Galveston, TXUSA
| |
Collapse
|
26
|
Greenwood TA, Light GA, Swerdlow NR, Calkins ME, Green MF, Gur RE, Gur RC, Lazzeroni LC, Nuechterlein KH, Olincy A, Radant AD, Seidman LJ, Siever LJ, Silverman JM, Stone WS, Sugar CA, Tsuang DW, Tsuang MT, Turetsky BI, Freedman R, Braff DL. Gating Deficit Heritability and Correlation With Increased Clinical Severity in Schizophrenia Patients With Positive Family History. Am J Psychiatry 2016; 173:385-91. [PMID: 26441157 PMCID: PMC4933520 DOI: 10.1176/appi.ajp.2015.15050605] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE The Consortium on the Genetics of Schizophrenia Family Study evaluated 12 primary and other supplementary neurocognitive and neurophysiological endophenotypes in schizophrenia probands and their families. Previous analyses of prepulse inhibition (PPI) and P50 gating measures in this sample revealed heritability estimates that were lower than expected based on earlier family studies. Here the authors investigated whether gating measures were more heritable in multiply affected families with a positive family history compared with families with only a single affected proband (singleton). METHOD A total of 296 nuclear families consisting of a schizophrenia proband, at least one unaffected sibling, and both parents underwent a comprehensive endophenotype and clinical characterization. The Family Interview for Genetic Studies was administered to all participants and used to obtain convergent psychiatric symptom information for additional first-degree relatives. Among the families, 97 were multiply affected, and 96 were singletons. RESULTS Both PPI and P50 gating displayed substantially increased heritability in the 97 multiply affected families (47% and 36%, respectively) compared with estimates derived from the entire sample of 296 families (29% and 20%, respectively). However, no evidence for heritability was observed for either measure in the 96 singleton families. Schizophrenia probands derived from the multiply affected families also displayed a significantly increased severity of clinical symptoms compared with those from singleton families. CONCLUSIONS PPI and P50 gating measures demonstrate substantially increased heritability in schizophrenia families with a higher genetic vulnerability for illness, providing further support for the commonality of genes underlying both schizophrenia and gating measures.
Collapse
Affiliation(s)
| | - Gregory A. Light
- Department of Psychiatry, University of California San Diego, La Jolla, CA,VISN-22 Mental Illness, Research, Education and Clinical Center (MIRECC), VA San Diego Healthcare System
| | - Neal R. Swerdlow
- Department of Psychiatry, University of California San Diego, La Jolla, CA
| | - Monica E. Calkins
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - Michael F. Green
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA,VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Raquel E. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - Ruben C. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - Laura C. Lazzeroni
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA
| | - Keith H. Nuechterlein
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA
| | - Ann Olincy
- Department of Psychiatry, University of Colorado Health Sciences Center, Denver, CO
| | - Allen D. Radant
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA,VA Puget Sound Health Care System, Seattle, WA
| | - Larry J. Seidman
- Department of Psychiatry, Harvard Medical School, Boston, MA,Massachusetts Mental Health Center Public Psychiatry Division of the Beth Israel Deaconess Medical Center, Boston, MA
| | - Larry J. Siever
- Department of Psychiatry, The Mount Sinai School of Medicine, New York, NY,James J. Peters VA Medical Center, New York, NY
| | - Jeremy M. Silverman
- Department of Psychiatry, The Mount Sinai School of Medicine, New York, NY,James J. Peters VA Medical Center, New York, NY
| | - William S. Stone
- Department of Psychiatry, Harvard Medical School, Boston, MA,Massachusetts Mental Health Center Public Psychiatry Division of the Beth Israel Deaconess Medical Center, Boston, MA
| | - Catherine A. Sugar
- Department of Biostatistics, University of California Los Angeles School of Public Health, Los Angeles, CA
| | - Debby W. Tsuang
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA,VA Puget Sound Health Care System, Seattle, WA
| | - Ming T. Tsuang
- Department of Psychiatry, University of California San Diego, La Jolla, CA,Center for Behavioral Genomics, and Institute for Genomic Medicine, University of California San Diego, La Jolla, CA,Harvard Institute of Psychiatric Epidemiology and Genetics, Boston, MA
| | - Bruce I. Turetsky
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - Robert Freedman
- Department of Psychiatry, University of Colorado Health Sciences Center, Denver, CO
| | - David L. Braff
- Department of Psychiatry, University of California San Diego, La Jolla, CA,VISN-22 Mental Illness, Research, Education and Clinical Center (MIRECC), VA San Diego Healthcare System
| |
Collapse
|
27
|
Memantine Effects On Sensorimotor Gating and Mismatch Negativity in Patients with Chronic Psychosis. Neuropsychopharmacology 2016; 41:419-30. [PMID: 26062785 PMCID: PMC5130118 DOI: 10.1038/npp.2015.162] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 04/23/2015] [Accepted: 05/14/2015] [Indexed: 12/17/2022]
Abstract
Patients with chronic psychotic disorders (CPD) exhibit deficient sensorimotor gating (measured by prepulse inhibition (PPI) of startle) and mismatch negativity (MMN). In healthy subjects (HS), N-methyl-D-aspartate (NMDA) antagonists like memantine and ketamine increase PPI, and under some conditions, memantine enhances MMN; these findings present a challenge to understanding the basis for deficient PPI and MMN in psychotic disorders, as reduced NMDA activity is implicated in the pathogenesis of these disorders. Here we assessed for the first time the effects of memantine on PPI and MMN in CPD subjects. Baseline PPI was measured in HS and patients with a diagnosis of schizophrenia or schizoaffective disorder, depressed type. Subjects (total n=84) were then tested twice, in a double-blind crossover design, comparing either: (1) placebo vs 10 mg of memantine or (2) placebo vs 20 mg memantine. Tests included measures of acoustic startle magnitude and habituation, PPI, MMN, autonomic indices, and subjective self-rating scales. Memantine (20 mg) significantly enhanced PPI in CPD subjects, and enhanced MMN across subject groups. These effects on PPI were age dependent and most evident in older CPD patients, whereas those on MMN were most evident in younger subjects. The lower dose (10 mg) either had no detectable effect or tended to degrade these measures. The NMDA antagonist, memantine, has dose-dependent effects on preconscious, automatic measures of sensorimotor gating and auditory sensory processing that are associated with enhanced cognition and function in CPD patients. Ongoing studies will determine whether these memantine-induced changes predict acute pro-cognitive or otherwise clinically beneficial effects in CPD patients.
Collapse
|
28
|
Bergeron SA, Carrier N, Li GH, Ahn S, Burgess HA. Gsx1 expression defines neurons required for prepulse inhibition. Mol Psychiatry 2015; 20:974-85. [PMID: 25224259 PMCID: PMC4362800 DOI: 10.1038/mp.2014.106] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 07/09/2014] [Accepted: 08/04/2014] [Indexed: 02/07/2023]
Abstract
In schizophrenia, cognitive overload is thought to reflect an inability to suppress non-salient information, a process which is studied using prepulse inhibition (PPI) of the startle response. PPI is reduced in schizophrenia and routinely tested in animal models and preclinical trials of antipsychotic drugs. However, the underlying neuronal circuitry is not well understood. We used a novel genetic screen in larval zebrafish to reveal the molecular identity of neurons that are required for PPI in fish and mice. Ablation or optogenetic silencing of neurons with developmental expression of the transcription factor genomic screen homeobox 1 (gsx1) produced profound defects in PPI in zebrafish, and PPI was similarly impaired in Gsx1 knockout mice. Gsx1-expressing neurons reside in the dorsal brainstem and form synapses closely apposed to neurons that initiate the startle response. Surprisingly, brainstem Gsx1 neurons are primarily glutamatergic despite their role in a functionally inhibitory pathway. As Gsx1 has an important role in regulating interneuron development in the forebrain, these findings reveal a molecular link between control of interneuron specification and circuits that gate sensory information across brain regions.
Collapse
Affiliation(s)
- Sadie A. Bergeron
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Nicole Carrier
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Grace H. Li
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Sohyun Ahn
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Harold A. Burgess
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA,6 Center Drive, Building 6B, Rm 3B308, Bethesda, MD 20892, , tel: 301-402-6018; fax: 301-496-0243
| |
Collapse
|
29
|
Bitanihirwe BKY, Woo TUW. Perineuronal nets and schizophrenia: the importance of neuronal coatings. Neurosci Biobehav Rev 2014; 45:85-99. [PMID: 24709070 DOI: 10.1016/j.neubiorev.2014.03.018] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 02/19/2014] [Accepted: 03/25/2014] [Indexed: 12/17/2022]
Abstract
Schizophrenia is a complex brain disorder associated with deficits in synaptic connectivity. The insidious onset of this illness during late adolescence and early adulthood has been reported to be dependent on several key processes of brain development including synaptic refinement, myelination and the physiological maturation of inhibitory neural networks. Interestingly, these events coincide with the appearance of perineuronal nets (PNNs), reticular structures composed of components of the extracellular matrix that coat a variety of cells in the mammalian brain. Until recently, the functions of the PNN had remained enigmatic, but are now considered to be important in development of the central nervous system, neuronal protection and synaptic plasticity, all elements which have been associated with schizophrenia. Here, we review the emerging evidence linking PNNs to schizophrenia. Future studies aimed at further elucidating the functions of PNNs will provide new insights into the pathophysiology of schizophrenia leading to the identification of novel therapeutic targets with the potential to restore normal synaptic integrity in the brain of patients afflicted by this illness.
Collapse
Affiliation(s)
| | - Tsung-Ung W Woo
- Program in Cellular Neuropathology, McLean Hospital, Belmont, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
30
|
Swerdlow NR, Light GA, Sprock J, Calkins ME, Green MF, Greenwood TA, Gur RE, Gur RC, Lazzeroni LC, Nuechterlein KH, Radant AD, Ray A, Seidman LJ, Siever LJ, Silverman JM, Stone WS, Sugar CA, Tsuang DW, Tsuang MT, Turetsky BI, Braff DL. Deficient prepulse inhibition in schizophrenia detected by the multi-site COGS. Schizophr Res 2014; 152:503-12. [PMID: 24405980 PMCID: PMC3960985 DOI: 10.1016/j.schres.2013.12.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 12/05/2013] [Accepted: 12/06/2013] [Indexed: 11/21/2022]
Abstract
BACKGROUND Startle inhibition by weak prepulses (PPI) is studied to understand the biology of information processing in schizophrenia patients and healthy comparison subjects (HCS). The Consortium on the Genetics of Schizophrenia (COGS) identified associations between PPI and single nucleotide polymorphisms in schizophrenia probands and unaffected relatives, and linkage analyses extended evidence for the genetics of PPI deficits in schizophrenia in the COGS-1 family study. These findings are being extended in a 5-site "COGS-2" study of 1800 patients and 1200 unrelated HCS to facilitate genetic analyses. We describe a planned interim analysis of COGS-2 PPI data. METHODS Eyeblink startle was measured in carefully screened HCS and schizophrenia patients (n=1402). Planned analyses of PPI (60 ms intervals) assessed effects of diagnosis, sex and test site, PPI-modifying effects of medications and smoking, and relationships between PPI and neurocognitive measures. RESULTS 884 subjects met strict inclusion criteria. ANOVA of PPI revealed significant effects of diagnosis (p=0.0005) and sex (p<0.002), and a significant diagnosis×test site interaction. HCS>schizophrenia PPI differences were greatest among patients not taking 2nd generation antipsychotics, and were independent of smoking status. Modest but significant relationships were detected between PPI and performance in specific neurocognitive measures. DISCUSSION The COGS-2 multi-site study detects schizophrenia-related PPI deficits reported in single-site studies, including patterns related to diagnosis, prepulse interval, sex, medication and other neurocognitive measures. Site differences were detected and explored. The target COGS-2 schizophrenia "endophenotype" of reduced PPI should prove valuable for identifying and confirming schizophrenia risk genes in future analyses.
Collapse
Affiliation(s)
- Neal R Swerdlow
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States.
| | - Gregory A Light
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States; VISN 22, Mental Illness Research, Education & Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, CA, United States
| | - Joyce Sprock
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States; VISN 22, Mental Illness Research, Education & Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, CA, United States
| | - Monica E Calkins
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael F Green
- Department of Psychiatry and Biobehavioral Sciences, Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States; VA Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Tiffany A Greenwood
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - Raquel E Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, United States
| | - Ruben C Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, United States
| | - Laura C Lazzeroni
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States; Department of Pediatrics, Stanford University, Stanford, CA, United States
| | - Keith H Nuechterlein
- Department of Psychiatry and Biobehavioral Sciences, Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Allen D Radant
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States; VA Puget Sound Health Care System, Seattle, WA, United States
| | - Amrita Ray
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States; Department of Pediatrics, Stanford University, Stanford, CA, United States
| | - Larry J Seidman
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States; Massachusetts Mental Health Center Public Psychiatry Division of the Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Larry J Siever
- Department of Psychiatry, The Mount Sinai School of Medicine, New York, NY, United States; James J. Peters VA Medical Center, New York, NY, United States
| | - Jeremy M Silverman
- Department of Psychiatry, The Mount Sinai School of Medicine, New York, NY, United States; James J. Peters VA Medical Center, New York, NY, United States
| | - William S Stone
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States; Massachusetts Mental Health Center Public Psychiatry Division of the Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Catherine A Sugar
- Department of Psychiatry and Biobehavioral Sciences, Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States; VISN 22, Mental Illness Research, Education & Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, CA, United States; Department of Biostatistics, University of California Los Angeles School of Public Health, Los Angeles, CA, United States
| | - Debby W Tsuang
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States; VA Puget Sound Health Care System, Seattle, WA, United States
| | - Ming T Tsuang
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States; Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, United States; Harvard Institute of Psychiatric Epidemiology and Genetics, Boston, MA, United States
| | - Bruce I Turetsky
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, United States
| | - David L Braff
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States; VISN 22, Mental Illness Research, Education & Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, CA, United States
| |
Collapse
|
31
|
Mohr C, Ettinger U. An Overview of the Association between Schizotypy and Dopamine. Front Psychiatry 2014; 5:184. [PMID: 25566103 PMCID: PMC4271513 DOI: 10.3389/fpsyt.2014.00184] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Accepted: 12/05/2014] [Indexed: 12/31/2022] Open
Abstract
Schizotypy refers to a constellation of personality traits that are believed to mirror the subclinical expression of schizophrenia in the general population. Evidence from pharmacological studies indicates that dopamine (DA) is involved in the etiology of schizophrenia. Based on the assumption of a continuum between schizophrenia and schizotypy, researchers have begun investigating the association between DA and schizotypy using a wide range of methods. In this article, we review published studies on this association from the following areas of work: (1) experimental investigations of the interactive effects of dopaminergic challenges and schizotypy on cognition, motor control, and behavior (2), dopaminergically supported cognitive functions (3), studies of associations between schizotypy and polymorphisms in genes involved in dopaminergic neurotransmission, and (4) molecular imaging studies of the association between schizotypy and markers of the DA system. Together, data from these lines of evidence suggest that DA is important to the expression and experience of schizotypy and associated behavioral biases. An important observation is that the experimental designs, methods, and manipulations used in this research are highly heterogeneous. Future studies are required to replicate individual observations, to enlighten the link between DA and different schizotypy dimensions (positive, negative, cognitive disorganization), and to guide the search for solid DA-sensitive behavioral markers. Such studies are important in order to clarify inconsistencies between studies. More work is also needed to identify differences between dopaminergic alterations in schizotypy compared to the dysfunctions observed in schizophrenia.
Collapse
Affiliation(s)
- Christine Mohr
- Institute of Psychology, University of Lausanne , Lausanne , Switzerland
| | - Ulrich Ettinger
- Department of Psychology, University of Bonn , Bonn , Germany
| |
Collapse
|
32
|
Disrupting the clustering of GABAA receptor α2 subunits in the frontal cortex leads to reduced γ-power and cognitive deficits. Proc Natl Acad Sci U S A 2013; 110:16628-33. [PMID: 24043839 DOI: 10.1073/pnas.1308706110] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In schizophrenia, cognitive dysfunction is highly predictive of poor patient outcomes and is not responsive to current medications. Postmortem studies have suggested that cognitive deficits in schizophrenia are correlated with modifications in the number and size of inhibitory synapses. To test if these modifications lead to cognitive deficits, we have created a dominant-negative virus [adeno-associated (AAV)-DN1] that disrupts the clustering of γ-aminobutyric acid type A receptors (GABA(A)Rs) at postsynaptic inhibitory specializations. When injected into the frontal cortex of mice, AAV-DN1 impairs GABA(A)R α2 subunit and GABA transporter 1 (GAT-1) clustering, but increases GABA(A)R α1 subunit clustering on the perisomatic region, with no influence on axon-initial segment clustering. Mice expressing AAV-DN1 have prepulse inhibition deficits and impairments in working memory. Significantly, these behavioral deficits are paralleled by a reduction in electroencephalography γ-power. Collectively, our study provides functional evidence revealing that GABAergic synapses in the prefrontal cortex directly contribute to cognition and γ-power.
Collapse
|
33
|
Abstract
In prepulse inhibition (PPI), the startle response to a strong, unexpected stimulus is diminished if shortly preceded by the onset of a different stimulus. Because deficits in this inhibitory gating process are a hallmark feature of schizophrenia and certain other psychiatric disorders, the mechanisms underlying PPI are of significant interest. We previously used the invertebrate model system Tritonia diomedea to identify the first cellular mechanism for PPI--presynaptic inhibition of transmitter release from the afferent neurons (S-cells) mediating the startle response. Here, we report the involvement of a second, more powerful PPI mechanism in Tritonia: prepulse-elicited conduction block of action potentials traveling in the startle pathway caused by identified inhibitory interneurons activated by the prepulse. This example of axo-axonic conduction block--neurons in one pathway inhibiting the propagation of action potentials in another--represents a novel and potent mechanism of sensory gating in prepulse inhibition.
Collapse
|
34
|
Ramage EM, Weintraub DM, Allen DN, Snyder JS. Evidence for stimulus-general impairments on auditory stream segregation tasks in schizophrenia. J Psychiatr Res 2012; 46:1540-5. [PMID: 23017812 PMCID: PMC3485434 DOI: 10.1016/j.jpsychires.2012.08.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 06/26/2012] [Accepted: 08/27/2012] [Indexed: 11/30/2022]
Abstract
BACKGROUND Auditory impairments in schizophrenia have been demonstrated previously, especially for tasks requiring precise encoding of frequency, although it is unclear the extent to which they have difficulty using pitch information and other cues to segregate sounds. We determined the extent to which those with schizophrenia have difficulty using pitch information and other auditory cues to segregate sounds that are presented sequentially. METHODS Ten participants with schizophrenia and nine healthy/normal control participants completed a battery of tasks that tested for the ability to perform sequential auditory stream segregation using pitch, amplitude modulation, or inter-aural phase difference as cues to segregation. RESULTS All three sequential segregation tasks showed reduced tendency for those with schizophrenia to perceive segregated sounds, compared to control participants. CONCLUSIONS These findings extend prior research by demonstrating a general impairment on sequential sound segregation tasks in schizophrenia, and not just on tasks that require precise encoding of frequency. Together, the pattern of results provide evidence that auditory impairments in schizophrenia result from selective abnormalities in neural circuits that carry out specific computations necessary for stream segregation, as opposed to an impairment in processing specific cues.
Collapse
Affiliation(s)
- Erin M Ramage
- Department of Psychology, University of Nevada, Las Vegas, NV 89154-5030, USA
| | | | | | | |
Collapse
|
35
|
Baisley SK, Fallace KL, Rajbhandari AK, Bakshi VP. Mutual independence of 5-HT(2) and α1 noradrenergic receptors in mediating deficits in sensorimotor gating. Psychopharmacology (Berl) 2012; 220:465-79. [PMID: 21947334 PMCID: PMC4090044 DOI: 10.1007/s00213-011-2490-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Accepted: 09/05/2011] [Indexed: 12/20/2022]
Abstract
RATIONALE Prepulse inhibition (PPI), a preattentional information-filtering mechanism, is disrupted by serotonin (5-HT) or norepinephrine (NE) agonists to model deficits seen in schizophrenia, but whether this effect occurs through interactions between these systems is not known. OBJECTIVES These studies investigated whether PPI/activity changes induced by agonists of one system were dependent on neurotransmission within the other. METHODS Male Sprague-Dawley rats received the 5-HT(2) receptor agonist DOI (1-[2,5-dimethoxy-4-iodophenyl]-2-aminopropane) (0, 0.3 mg/kg), with or without antagonists for α1 (prazosin:0, 0.3, or 1 mg/kg) or β (timolol:0, 3, or 10 mg/kg) receptors or their combination (0 or 0.3 mg/kg prazosin + 3 mg/kg timolol), or the 5-HT(2) antagonist ritanserin (0, 2 mg/kg). Separately, the α1-adrenergic receptor agonist cirazoline (0, 0.68 mg/kg) was given with and without ritanserin (0, 0.5, or 2 mg/kg) or the NE antagonists (0 or 0.3 mg/kg prazosin + 3 mg/kg timolol). Finally, combinations of subthreshold doses of DOI (0, 0.01, 0.025 mg/kg) and cirazoline (0, 0.1, 0.25 mg/kg) were tested for their ability to disrupt PPI, and concomitant administration of all three antagonists (0 vs. 0.3 mg/kg prazosin + 3 mg/kg timolol + 2 mg/kg ritanserin) was assessed for its ability to modify PPI. Locomotion was assessed in an additional set of experiments. RESULTS Doses/combinations of prazosin and timolol that reversed cirazoline-induced effects did not alter DOI-induced effects, and ritanserin did not affect cirazoline at doses that blocked DOI-mediated effects. Concomitant antagonism of α1 + β + 5-HT(2) receptors did not modify PPI, nor did combinations of subthreshold doses of cirazoline and DOI. CONCLUSIONS 5-HT(2) receptors and α1 and β NE receptors may act through independent mechanisms to modulate sensorimotor gating and locomotor activity.
Collapse
Affiliation(s)
- Sarah K. Baisley
- Department of Psychiatry, UW-Madison, Madison, WI, USA,Neuroscience Training Program, UW-Madison, Madison, WI, USA
| | | | - Abha K. Rajbhandari
- Department of Psychiatry, UW-Madison, Madison, WI, USA,Neuroscience Training Program, UW-Madison, Madison, WI, USA
| | - Vaishali P. Bakshi
- Department of Psychiatry, UW-Madison, Madison, WI, USA,Neuroscience Training Program, UW-Madison, Madison, WI, USA,Department of Psychiatry, University of Wisconsin School of Medicine and Public Health, 6001 Research Park Blvd, Madison, WI 53719, USA
| |
Collapse
|
36
|
Valsamis B, Schmid S. Habituation and prepulse inhibition of acoustic startle in rodents. J Vis Exp 2011:e3446. [PMID: 21912367 DOI: 10.3791/3446] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The acoustic startle response is a protective response, elicited by a sudden and intense acoustic stimulus. Facial and skeletal muscles are activated within a few milliseconds, leading to a whole body flinch in rodents(1). Although startle responses are reflexive responses that can be reliably elicited, they are not stereotypic. They can be modulated by emotions such as fear (fear potentiated startle) and joy (joy attenuated startle), by non-associative learning processes such as habituation and sensitization, and by other sensory stimuli through sensory gating processes (prepulse inhibition), turning startle responses into an excellent tool for assessing emotions, learning, and sensory gating, for review see( 2, 3). The primary pathway mediating startle responses is very short and well described, qualifying startle also as an excellent model for studying the underlying mechanisms for behavioural plasticity on a cellular/molecular level(3). We here describe a method for assessing short-term habituation, long-term habituation and prepulse inhibition of acoustic startle responses in rodents. Habituation describes the decrease of the startle response magnitude upon repeated presentation of the same stimulus. Habituation within a testing session is called short-term habituation (STH) and is reversible upon a period of several minutes without stimulation. Habituation between testing sessions is called long-term habituation (LTH)(4). Habituation is stimulus specific(5). Prepulse inhibition is the attenuation of a startle response by a preceding non-startling sensory stimulus(6). The interval between prepulse and startle stimulus can vary from 6 to up to 2000 ms. The prepulse can be any modality, however, acoustic prepulses are the most commonly used. Habituation is a form of non-associative learning. It can also be viewed as a form of sensory filtering, since it reduces the organisms' response to a non-threatening stimulus. Prepulse inhibition (PPI) was originally developed in human neuropsychiatric research as an operational measure for sensory gating(7). PPI deficits may represent the interface of "psychosis and cognition" as they seem to predict cognitive impairment(8-10). Both habituation and PPI are disrupted in patients suffering from schizophrenia(11), and PPI disruptions have shown to be, at least in some cases, amenable to treatment with mostly atypical antipsychotics(12, 13). However, other mental and neurodegenerative diseases are also accompanied by disruption in habituation and/or PPI, such as autism spectrum disorders (slower habituation), obsessive compulsive disorder, Tourette's syndrome, Huntington's disease, Parkinson's disease, and Alzheimer's Disease (PPI)(11, 14, 15) Dopamine induced PPI deficits are a commonly used animal model for the screening of antipsychotic drugs(16), but PPI deficits can also be induced by many other psychomimetic drugs, environmental modifications and surgical procedures.
Collapse
Affiliation(s)
- Bridget Valsamis
- Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario
| | | |
Collapse
|
37
|
Hasenkamp W, Kelley M, Egan G, Green A, Wilcox L, Boshoven W, Lewison B, Duncan E. Lack of relationship between acoustic startle and cognitive variables in schizophrenia and control subjects. Psychiatry Res 2011; 187:324-8. [PMID: 21397338 PMCID: PMC3078961 DOI: 10.1016/j.psychres.2011.02.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Revised: 02/07/2011] [Accepted: 02/08/2011] [Indexed: 11/19/2022]
Abstract
Measures of acoustic startle such as prepulse inhibition (PPI) and startle latency have been found to be impaired in schizophrenia, and are commonly thought to be related to cognitive deficits in this disease. However, findings about the relationship between startle variables and cognitive performance have been equivocal. In this study, we examined correlations between startle measures (baseline startle magnitude, latency, habituation and PPI) and cognitive performance (using the Benton Visual Retention Test, Conner's Continuous Performance Test, California Verbal Learning Test, Finger Tapping Test, and Wisconsin Card Sort Test) in 107 schizophrenia patients and 94 healthy controls. Overall, there was a lack of any significant relationship between these constructs in both populations when correcting for multiple comparisons. This suggests that alterations in startle measures seen in schizophrenia may not reflect elements of information processing that cause cognitive deficits in the disease.
Collapse
Affiliation(s)
- Wendy Hasenkamp
- Mental Health Service, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Heritability of acoustic startle magnitude, prepulse inhibition, and startle latency in schizophrenia and control families. Psychiatry Res 2010; 178:236-43. [PMID: 20483176 PMCID: PMC2902662 DOI: 10.1016/j.psychres.2009.11.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Revised: 10/29/2009] [Accepted: 11/09/2009] [Indexed: 11/20/2022]
Abstract
Prepulse inhibition (PPI) is an acoustic startle paradigm that has been used as an operational measure of sensorimotor gating. Many patients with schizophrenia have impaired PPI, and several lines of evidence suggest that PPI may represent a heritable endophenotype in this disease. We examined startle magnitude and latencies in 40 schizophrenia patients, 58 first-degree relatives of these patients, and 100 healthy controls. After removing low-startlers, we investigated PPI and startle habituation in 34 schizophrenia patients, 43 relatives, and 86 control subjects. Heritability analyses were conducted using a variance-component approach. We found significant heritability of 45% for PPI at the 60-ms interval and 67% for startle magnitude. Onset latency heritability estimates ranged between 39% and 90% across trial types, and those for peak latency ranged from 29% to 68%. Heritability of startle habituation trended toward significance at 31%. We did not detect differences between controls and either schizophrenia patients or their family members for PPI, startle magnitude, or habituation. Startle latencies were generally longer in schizophrenia patients than controls. The heritability findings give impetus to applying genetic analyses to PPI variables, and suggest that startle latency may also be a useful measure in the study of potential endophenotypes for schizophrenia.
Collapse
|
39
|
Swerdlow NR, Talledo JA. Effects of the first prepulse on the blink response to a startling noise. Behav Neurosci 2009; 123:607-13. [PMID: 19485567 PMCID: PMC5944322 DOI: 10.1037/a0015064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Startle is inhibited when a startling stimulus follows 30-300 ms after a weak prepulse. Prepulse inhibition (PPI) is an operational measure of sensorimotor gating and is deficient in several neuropsychiatric disorders. Previous reports argue both for and against a learned component to the inhibitory effects of prepulses, but this issue has yet to be fully investigated using stimuli that most commonly detect PPI deficits in clinical populations. If the inhibitory impact of a prepulse is learned, PPI should not be evident when the prepulse is the first stimulus experienced by the subject. Eyeblink electromyography in normal adults was recorded after either a 118 dB(A) 40-ms noise pulse alone (PA) or the same pulse preceded 120 ms by an 86 dB(A) 5-ms noise prepulse (pp+P). In 25 subjects (Order 1), Trial 1 was a PA, and Trial 2 was a pp+P; 23 subjects experienced the opposite order (Order 2). In 34 subjects, Trials 1 and 2 were both PA (control order). Background was 70 dB(A). Startle magnitude increased from Trial 1 to 2 if no prepulse was presented (control order). Compared with the control order, startle inhibition by prepulses was evident in both Orders 1 and 2, and was more robust in Order 2 (first trial=pp+P). Startle magnitude was significantly lower on pp+P than on PA trials in Order 2 but not Order 1 (F<1). Prepulses inhibit startle on the first pairing with a startling pulse, an effect that cannot be explained by learning.
Collapse
Affiliation(s)
- Neal R Swerdlow
- Department of Psychiatry, University of California, San Diego, School of Medicine, La Jolla, CA 92093-0804, USA.
| | | |
Collapse
|
40
|
Allen AJ, Griss ME, Folley BS, Hawkins KA, Pearlson GD. Endophenotypes in schizophrenia: a selective review. Schizophr Res 2009; 109:24-37. [PMID: 19223268 PMCID: PMC2665704 DOI: 10.1016/j.schres.2009.01.016] [Citation(s) in RCA: 156] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2008] [Revised: 01/11/2009] [Accepted: 01/14/2009] [Indexed: 10/21/2022]
Abstract
BACKGROUND Given the wealth of data in the literature on schizophrenia endophenotypes, it is useful to have one source to reference their frequency data. We reviewed the literature on disease-liability associated variants in structural and functional magnetic resonance images (MRI), sensory processing measures, neuromotor abilities, neuropsychological measures, and physical characteristics in schizophrenia patients (SCZ), their first-degree relatives (REL), and healthy controls (HC). The purpose of this review was to provide a summary of the existing data on the most extensively published endophenotypes for schizophrenia. METHODS We searched PubMed and MedLine for all studies on schizophrenia endophenotypes comparing SCZ to HC and/or REL to HC groups. Percent abnormal values, generally defined as >2 SD from the mean (in the direction of abnormality) and/or associated effect sizes (Cohen's d) were calculated for each study. RESULTS Combined, the articles reported an average 39.4% (SD=20.7%; range=2.2-100%) of abnormal values in SCZ, 28.1% (SD=16.6%; range=1.6-67.0%) abnormal values in REL, and 10.2% (SD=6.7%; range=0.0-34.6%) in HC groups. CONCLUSIONS These findings are reviewed in the context of emerging hypotheses on schizophrenia endophenotypes, as well as a discussion of clustering trends among the various intermediate phenotypes. In addition, programs for future research are discussed, as instantiated in a few recent large-scale studies on multiple endophenotypes across patients, relatives, and healthy controls.
Collapse
Affiliation(s)
- Allyssa J. Allen
- Olin Neuropsychiatry Research Center, Institute of Living at Hartford Hospital, 200 Retreat Avenue, Hartford, CT 06106,Corresponding Author: Allyssa J. Allen, Olin Neuropsychiatry Research Center, Whitehall Building, 200 Retreat Avenue, Hartford, CT 06106, Tel: 860-459-7806, Fax: 860-545-7797,
| | - Mélina E. Griss
- Olin Neuropsychiatry Research Center, Institute of Living at Hartford Hospital, 200 Retreat Avenue, Hartford, CT 06106
| | - Bradley S. Folley
- Olin Neuropsychiatry Research Center, Institute of Living at Hartford Hospital, 200 Retreat Avenue, Hartford, CT 06106
| | - Keith A. Hawkins
- Dept. of Psychiatry, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06511
| | - Godfrey D. Pearlson
- Olin Neuropsychiatry Research Center, Institute of Living at Hartford Hospital, 200 Retreat Avenue, Hartford, CT 06106,Dept. of Psychiatry, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06511
| |
Collapse
|
41
|
Turetsky BI, Bilker WB, Siegel SJ, Kohler CG, Gur RE. Profile of auditory information-processing deficits in schizophrenia. Psychiatry Res 2009; 165:27-37. [PMID: 18990453 PMCID: PMC2652872 DOI: 10.1016/j.psychres.2008.04.013] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2007] [Revised: 01/16/2008] [Accepted: 04/14/2008] [Indexed: 10/21/2022]
Abstract
Schizophrenia patients exhibit abnormalities in several different auditory event-related potential (ERP) measures. It is unclear how these abnormalities relate to each other, since multiple measures are rarely acquired from the same sample. This study addressed two related questions: 1) Are specific auditory ERP measures differentially impaired in schizophrenia? 2) Do abnormalities co-aggregate within the same patients? Nine auditory ERP measures were acquired in a single testing session from 23 schizophrenia patients and 22 healthy subjects. Hierarchical oblique factor analysis revealed that these measures aggregated into four factors, with each loading primarily on a single factor. Patient deficits were observed for two independent factors: N100/mismatch negativity (MMN) and P3a/P3b. N100/MMN abnormalities were associated with symptoms of alogia and formal thought disorder. P3a/P3b abnormalities were associated with avolition, attentional disturbances and delusions. We conclude that deficits in different ERP measures of early sensory processing at the level of the auditory cortex co-occur in patients. These likely represent a single differential deficit indexing the physiological abnormality underlying impaired language and verbal processing. This is relatively independent of a higher cortical deficit that mediates cognitive stimulus evaluation and underlies deficits in motivation, attention and reality testing. Such multidimensional profiling of ERP abnormalities may help to clarify the clinical and genetic heterogeneity of schizophrenia.
Collapse
Affiliation(s)
- Bruce I. Turetsky
- Department of Psychiatry, University of Pennsylvania Philadelphia, PA, USA,Correspondence: Bruce I. Turetsky, M.D., Neuropsychiatry Division, Department of Psychiatry, 10th Floor, Gates Building, University of Pennsylvania School of Medicine, 3400 Spruce Street, Philadelphia, PA 19104. Tel: (215) 615-3607. Fax: (215) 662-7903., E-mail:
| | - Warren B. Bilker
- Department Biostatistics and Epidemiology, University of Pennsylvania Philadelphia, PA, USA
| | - Steven J. Siegel
- Department of Psychiatry, University of Pennsylvania Philadelphia, PA, USA
| | | | - Raquel E. Gur
- Department of Psychiatry, University of Pennsylvania Philadelphia, PA, USA
| |
Collapse
|
42
|
Geyer MA. Developing translational animal models for symptoms of schizophrenia or bipolar mania. Neurotox Res 2009; 14:71-8. [PMID: 18790726 DOI: 10.1007/bf03033576] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Animal models have long been used to explore hypotheses regarding the neurobiological substrates of and treatments for psychiatric disorders. Early attempts to develop models that mimic the entirety of the diagnostic syndromes in psychiatry have evolved into more appropriate efforts to model specific symptoms. Such an approach reflects the facts that even in patients, clinical symptoms transcend diagnostic categories, and the specific etiologies of psychiatric disorders are unknown. An animal model can only be identified adequately by specifying both the manipulation (drug, lesion, strain) used to induce abnormalities and the measure(s) used to characterize them. A wide range of pharmacological, lesion, and developmental manipulations have been combined with various measures of information processing to develop useful animal models that parallel human tests. Prepulse inhibition of startle, event-related potential (ERP) measures of auditory gating, and Cambridge neuropsychological test automated battery (CANTAB) measures of cognition are examples of measures that can be used in both rodents and humans and that are robustly altered in both psychiatric disorders and animals manipulated with appropriate drugs or lesions. The further development of cross-species models is critically important, given the new opportunities for the development and registration of specific treatments for the cognitive disorders of schizophrenia that are not ameliorated by available drugs. In moving beyond the focus on psychotic symptoms to the cognitive symptoms of schizophrenia, animal models that do not involve manipulations of dopamine D(2) receptors but that do utilize information processing measures that are correlated with cognitive disturbances are receiving increased attention. Here, selected examples of how cross-species measures of psychiatric disorders are developed and validated are discussed. Specific animal paradigms that parallel the specific domains of cognition that are altered in schizophrenia provide one focus of the review. Another focus includes efforts to develop new human models of psychiatric symptoms that are designed to parallel existing tests used in rodents.
Collapse
Affiliation(s)
- M A Geyer
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093-0804, USA.
| |
Collapse
|
43
|
Csomor PA, Yee BK, Feldon J, Theodoridou A, Studerus E, Vollenweider FX. Impaired prepulse inhibition and prepulse-elicited reactivity but intact reflex circuit excitability in unmedicated schizophrenia patients: a comparison with healthy subjects and medicated schizophrenia patients. Schizophr Bull 2009; 35:244-55. [PMID: 18245063 PMCID: PMC2643951 DOI: 10.1093/schbul/sbm146] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Deficient sensorimotor gating as indexed by prepulse inhibition (PPI) of the startle response has been reported repeatedly in patients suffering from schizophrenia. According to the widely accepted "protective hypothesis," PPI reflects the protection of ongoing information processing against interference by other stimuli. Alternatively, it has been proposed that PPI might be regulated by startle reflex circuit excitability. In the present study, we evaluated these 2 conceptually divergent approaches underlying the regulation of PPI. To this end, we assessed sensorimotor gating as indexed by PPI, the reactivity to the prepulse-alone stimulus indexed as prepulse-elicited reactivity (PPER), and acoustic blink reflex excitability in terms of paired pulse suppression (PPS) within a single recording session in 13 unmedicated and 24 medicated (11 first break) schizophrenia patients in comparison to 43 healthy control subjects. The results showed that PPI was significantly reduced in unmedicated, but not in medicated schizophrenia patients. Furthermore, unmedicated patients could be distinguished from the medicated patients and control subjects in terms of PPER. In contrast to PPI, PPS did not differ between patients and control subjects. These findings are in line with the "protective hypothesis" of PPI and indicate that reduced sensorimotor gating in schizophrenia patients might be based on a reduced perception and/or processing of the prepulse stimulus. The extent to which PPER may or may not be causally associated with sensorimotor gating in schizophrenia has to be further investigated in human and animal studies.
Collapse
Affiliation(s)
- Philipp A. Csomor
- University Hospital of Psychiatry Zurich, PO Box 1931, CH-8032 Zurich, Switzerland,Laboratory of Behavioural Neurobiology, ETH Zurich, Schorenstrasse 16, CH-8603 Schwerzenbach, Switzerland,To whom correspondence should be addressed; tel: +41-44-3822407, fax: +41-44-384-2499, e-mail:
| | - Benjamin K. Yee
- Laboratory of Behavioural Neurobiology, ETH Zurich, Schorenstrasse 16, CH-8603 Schwerzenbach, Switzerland
| | - Joram Feldon
- Laboratory of Behavioural Neurobiology, ETH Zurich, Schorenstrasse 16, CH-8603 Schwerzenbach, Switzerland
| | | | - Erich Studerus
- Psychiatric Services of Aargau Canton, Department of Research, PO Box 298, CH-5201 Brugg, Switzerland
| | | |
Collapse
|
44
|
Atypical antipsychotics clozapine and quetiapine attenuate prepulse inhibition deficits in dopamine transporter knockout mice. Behav Pharmacol 2008; 19:562-5. [PMID: 18690110 DOI: 10.1097/fbp.0b013e32830dc110] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Sensorimotor gating disruptions are seen in various psychiatric illnesses with putatively different pathologies, including schizophrenia and bipolar disorder. Interestingly, mice lacking the dopamine (DA) transporter (DAT) gene display markedly increased levels of DA, deficits in sensorimotor gating, and hyperactivity relative to wild-type mice. Atypical antipsychotics are effective treatments of schizophrenia and manic symptoms, presumably in part by antagonizing DA receptors. Here we report that treatment with clozapine (3 mg/kg) or quetiapine (2.5 mg/kg) attenuated prepulse inhibition deficits in male DAT knockout mice. Thus male DAT knockout mice may provide a useful animal model for predicting the efficacy of novel drugs in treating psychiatric illnesses characterized by a dysregulated DA system.
Collapse
|
45
|
Hazlett EA, Buchsbaum MS, Zhang J, Newmark RE, Glanton CF, Zelmanova Y, Haznedar MM, Chu KW, Nenadic I, Kemether EM, Tang CY, New AS, Siever LJ. Frontal-striatal-thalamic mediodorsal nucleus dysfunction in schizophrenia-spectrum patients during sensorimotor gating. Neuroimage 2008; 42:1164-77. [PMID: 18588988 PMCID: PMC2548278 DOI: 10.1016/j.neuroimage.2008.05.039] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 05/15/2008] [Accepted: 05/19/2008] [Indexed: 10/22/2022] Open
Abstract
Prepulse inhibition (PPI) refers to a reduction in the amplitude of the startle eyeblink reflex to a strong sensory stimulus, the pulse, when it is preceded shortly by a weak stimulus, the prepulse. PPI is a measure of sensorimotor gating which serves to prevent the interruption of early attentional processing and it is impaired in schizophrenia-spectrum patients. In healthy individuals, PPI is more robust when attending to than ignoring a prepulse. Animal and human work demonstrates that frontal-striatal-thalamic (FST) circuitry modulates PPI. This study used functional magnetic resonance imaging (fMRI) to investigate FST circuitry during an attention-to-prepulse paradigm in 26 unmedicated schizophrenia-spectrum patients (13 schizotypal personality disorder (SPD), 13 schizophrenia) and 13 healthy controls. During 3T-fMRI acquisition and separately measured psychophysiological assessment of PPI, participants heard an intermixed series of high- and low-pitched tones serving as prepulses to an acoustic-startle stimulus. Event-related BOLD response amplitude curves in FST regions traced on co-registered anatomical MRI were examined. Controls showed greater activation during attended than ignored PPI conditions in all FST regions-dorsolateral prefrontal cortex (Brodmann areas 46, 9), striatum (caudate, putamen), and the thalamic mediodorsal nucleus. In contrast, schizophrenia patients failed to show differential BOLD responses in FST circuitry during attended and ignored prepulses, whereas SPD patients showed greater-than-normal activation during ignored prepulses. Among the three diagnostic groups, lower left caudate BOLD activation during the attended PPI condition was associated with more deficient sensorimotor gating as measured by PPI. Schizophrenia-spectrum patients exhibit inefficient utilization of FST circuitry during attentional modulation of PPI. Schizophrenia patients have reduced recruitment of FST circuitry during task-relevant stimuli, whereas SPD patients allocate excessive resources during task-irrelevant stimuli. Dysfunctional FST activation, particularly in the caudate may underlie PPI abnormalities in schizophrenia-spectrum patients.
Collapse
Affiliation(s)
- Erin A Hazlett
- Department of Psychiatry, Box 1505, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Kumari V, Peters ER, Fannon D, Premkumar P, Aasen I, Cooke MA, Anilkumar AP, Kuipers E. Uncontrollable voices and their relationship to gating deficits in schizophrenia. Schizophr Res 2008; 101:185-94. [PMID: 18262774 PMCID: PMC2845800 DOI: 10.1016/j.schres.2007.12.481] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2007] [Revised: 12/06/2007] [Accepted: 12/23/2007] [Indexed: 11/16/2022]
Abstract
BACKGROUND Prepulse inhibition (PPI) of the startle response refers to the ability of a weak prestimulus to transiently inhibit the response to a closely following strong sensory stimulus. This effect is reduced in a number of disorders known to be associated with impaired gating of sensory, cognitive or motor information. The aim of this study was to investigate PPI deficit in relation to the dimensions of auditory hallucinations in patients with schizophrenia or schizoaffective disorder. METHOD PPI of the acoustically elicited eye blink startle response was measured electromyographically in 62 patients with schizophrenia (n=55) or schizoaffective disorder (n=7) (26 of 62 with current auditory hallucinations) and 22 healthy participants matched, on average, to age and sex of the patient group. RESULTS Patients, as a group, showed reduced PPI compared to healthy participants. The presence of auditory hallucinations was associated with a marked PPI deficit if the patients felt that they had no control over their occurrence and that they were unable to dismiss them. Hearing voices with a high degree of negative content was associated with high mean startle amplitude in patients with current auditory hallucinations. CONCLUSIONS Although auditory hallucinations in patients with schizophrenia are theorised to result from impaired monitoring of inner speech, the inability to consciously ignore them appears to be associated with a gating deficit. Hearing voices with negative content is associated with hyper-startle responding, possibly because such voices are threatening and thus provoke anxiety.
Collapse
Affiliation(s)
- Veena Kumari
- Department of Psychology, Institute of Psychiatry, King's College London, London, UK.
| | - Emmanuelle R. Peters
- Department of Psychology, Institute of Psychiatry, King's College London, London, UK
| | - Dominic Fannon
- Division of Psychological Medicine and Psychiatry, Institute of Psychiatry, King's College London, London, UK
| | - Preethi Premkumar
- Department of Psychology, Institute of Psychiatry, King's College London, London, UK
| | - Ingrid Aasen
- Department of Psychology, Institute of Psychiatry, King's College London, London, UK
| | - Michael A. Cooke
- Department of Psychology, Institute of Psychiatry, King's College London, London, UK
| | - Anantha P. Anilkumar
- Division of Psychological Medicine and Psychiatry, Institute of Psychiatry, King's College London, London, UK
| | - Elizabeth Kuipers
- Department of Psychology, Institute of Psychiatry, King's College London, London, UK
| |
Collapse
|
47
|
Wynn JK, Green MF, Sprock J, Light GA, Widmark C, Reist C, Erhart S, Marder SR, Mintz J, Braff DL. Effects of olanzapine, risperidone and haloperidol on prepulse inhibition in schizophrenia patients: a double-blind, randomized controlled trial. Schizophr Res 2007; 95:134-42. [PMID: 17662577 PMCID: PMC2716219 DOI: 10.1016/j.schres.2007.05.039] [Citation(s) in RCA: 59] [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: 03/02/2007] [Revised: 05/25/2007] [Accepted: 05/31/2007] [Indexed: 11/25/2022]
Abstract
Prepulse inhibition (PPI), whereby the startle eyeblink response is inhibited by a relatively weak non-startling stimulus preceding the powerful startle eliciting stimulus, is a measure of sensorimotor gating and has been shown to be deficient in schizophrenia patients. There is considerable interest in whether conventional and/or atypical antipsychotic medications can "normalize" PPI deficits in schizophrenia patients. 51 schizophrenia patients participated in a randomized, double-blind controlled trial on the effects of three commonly-prescribed antipsychotic medications (risperidone, olanzapine, or haloperidol) on PPI, startle habituation, and startle reactivity. Patients were tested at baseline, Week 4 and Week 8. Mixed model regression analyses revealed that olanzapine significantly improved PPI from Week 4 to Week 8, and that at Week 8 patients receiving olanzapine produced significantly greater PPI than those receiving risperidone, but not haloperidol. There were no effects of medication on startle habituation or startle reactivity. These results support the conclusion that olanzapine effectively increased PPI in schizophrenia patients, but that risperidone and haloperidol had no such effects. The results are discussed in terms of animal models, neural substrates, and treatment implications.
Collapse
Affiliation(s)
- Jonathan K Wynn
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Control of behavior in the natural environment where sensory stimuli are abundant requires superfluous information to be ignored. In part, this is achieved through selective transmission, or gating of signals to motor systems. A quantitative and clinically important measure of sensorimotor gating is prepulse inhibition (PPI) of the startle response, impairments in which have been demonstrated in several neuropsychiatric disorders, including schizophrenia. Here, we show for the first time that the acoustic startle response in zebrafish larvae is modulated by weak prepulses in a manner similar to mammalian PPI. We demonstrate that, like in mammals, antipsychotic drugs can suppress disruptions in zebrafish PPI induced by dopamine agonists. Because genetic factors underlying PPI are not well understood, we performed a screen and isolated mutant lines with reduced PPI. Analysis of Ophelia mutants demonstrates that they have normal sensory acuity and startle performance, but reduced PPI, suggesting that Ophelia is critical for central processing of sensory information. Thus, our results provide the first evidence for sensorimotor gating in larval zebrafish and report on the first unbiased screen to identify genes regulating this process.
Collapse
Affiliation(s)
- Harold A. Burgess
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-6058
| | - Michael Granato
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-6058
| |
Collapse
|
49
|
Abstract
Prepulse inhibition (PPI) of startle is an operational measure of the pre-attentive filtering process known as sensorimotor gating. Originally identified in patients with schizophrenia, PPI deficits have been observed in multiple but not all psychiatric disorders. Thus, as with most signs and symptoms of psychiatric disorders, deficits in PPI cut across diagnostic categories. It remains unclear whether the diversity of disorders exhibiting deficient PPI bespeaks diagnostic overlaps or comorbidities. Given the recent focus on treatments for cognitive deficits of schizophrenia independently of treating psychosis, the relationship of PPI deficits to cognitive deficits becomes of interest. Although PPI cannot be considered to be a cognitive process per se, abnormalities in pre-attentive information processing may be predictive of or lead to complex cognitive deficits. Animal models of PPI deficits produced by dopamine agonists reliably predict existing antipsychotics. Nevertheless, since neither PPI nor cognitive deficits in schizophrenia are ameliorated by standard antipsychotics, current research is exploring the predictive value of non-dopaminergic PPI models in identifying treatments for gating disturbances independently of their relevance to specific disorders. Both PPI and cognitive deficits in schizophrenia patients are not reversed by first generation antipsychotics but may be attenuated by clozapine. Similarly, effects of glutamate antagonists on symptoms in patients and PPI in animals appear to be reduced by clozapine. Hence, treatment-induced reversals of deficits in PPI produced by glutamate antagonists may provide animal, and human, models to aid in the discovery of treatments of cognitive deficits in patients already treated with existing antipsychotics.
Collapse
Affiliation(s)
- Mark A Geyer
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093-0804, USA.
| |
Collapse
|
50
|
Homayoun H, Moghaddam B. Fine-tuning of awake prefrontal cortex neurons by clozapine: comparison with haloperidol and N-desmethylclozapine. Biol Psychiatry 2007; 61:679-87. [PMID: 17046721 PMCID: PMC2910426 DOI: 10.1016/j.biopsych.2006.05.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2006] [Revised: 05/17/2006] [Accepted: 05/22/2006] [Indexed: 10/23/2022]
Abstract
BACKGROUND Mechanisms underlying clozapine's better clinical efficacy in schizophrenia remain poorly understood. The prefrontal cortex (PFC) has been implicated as a primary site for the therapeutic effects of clozapine; however, evidence for how clozapine influences the activity of PFC neurons in behaviorally relevant contexts is lacking. METHODS Ensemble single unit recording in awake rats was used to measure the activity of PFC neurons in response to clozapine, its main metabolite N-desmethylclozapine (DMClz), and the typical antipsychotic drug haloperidol during baseline conditions and after treatment with the N-methyl-D-aspartate antagonist MK801. Behavioral stereotypy was scored during recording. RESULTS Clozapine and DMClz but not haloperidol had an activity-dependent influence on spontaneous firing rate of PFC cells: they increased the activity of neurons with low baseline firing rates and decreased the activity of neurons with higher firing rates. Clozapine and DMClz but not haloperidol also reversed the effect of MK801 on PFC neuronal firing. This reversal was strongly correlated with blockade of MK801-induced behavioral stereotypy. CONCLUSIONS These findings indicate that clozapine has the capacity to fine-tune spontaneous and disrupted activity of PFC neurons. This effect might contribute, in part, to the therapeutic efficacy of clozapine in schizophrenia.
Collapse
Affiliation(s)
- Houman Homayoun
- Department of Neurosciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|