1
|
Hu Y, Liu W, Wu Y, Hu Z, Tao Y, Zhang Q, Chen J, Li M, Hu L, Ding Y. DCC in the cerebral cortex is required for cognitive functions in mouse. Brain Pathol 2025; 35:e13306. [PMID: 39293934 PMCID: PMC11961207 DOI: 10.1111/bpa.13306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/26/2024] [Indexed: 09/20/2024] Open
Abstract
Schizophrenia (SZ) is a highly heritable mental disorder, and genome-wide association studies have identified the association between deleted in colorectal cancer (DCC) and SZ. Previous study has shown a lowered expression of DCC in the cerebral cortex of SZ patient. In this study, we identified novel single nucleotide polymorphisms (SNPs) of DCC statistically correlated with SZ. Based on these, we generated DCC conditional knockout (CKO) mice and explored behavioral phenotypes in these mice. We observed that deletion of DCC in cortical layer VI but not layer V led to deficits in fear and spatial memory, as well as defective sensorimotor gating revealed by the prepulse inhibition test (PPI). Critically, the defective sensorimotor gating could be restored by olanzapine, an antipsychotic drug. Furthermore, we found that the levels of p-AKT and p-GSK3α/β were decreased, which was responsible for impaired PPI in the DCC-deficient mice. Finally, the DCC-deficient mice also displayed reduced spine density of pyramidal neurons and disturbed delta-oscillations. Our data, for the first time, identified and explored downstream substrates and signaling pathway of DCC which supports the hypothesis that DCC is a SZ-related risky gene and when defective, may promote SZ-like pathogenesis and behavioral phenotypes in mice.
Collapse
Affiliation(s)
- Yun‐Qing Hu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Wei‐Tang Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
- Shanghai Institute of Infectious Disease and BiosecurityFudan UniversityShanghaiChina
| | - Yong Wu
- Research Center for Mental Health and Neuroscience, Wuhan Mental Health CenterJianghan UniversityWuhanChina
| | - Zhi‐Bin Hu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Yun‐Chao Tao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Qiong Zhang
- Department of Laboratory Animal ScienceFudan UniversityShanghaiChina
| | - Jia‐Yin Chen
- Department of Laboratory Animal ScienceFudan UniversityShanghaiChina
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of ZoologyChinese Academy of SciencesKunmingChina
| | - Ling Hu
- Department of Laboratory Animal ScienceFudan UniversityShanghaiChina
| | - Yu‐Qiang Ding
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
- Department of Laboratory Animal ScienceFudan UniversityShanghaiChina
| |
Collapse
|
2
|
Zhou H, Liu P, Zhang C, Xue X, Chen X, Zhang Z, Shen W, Yang S, Fan M, Wang F. Gap-induced inhibition of the post-auricular muscle response in miniature pigs. Acta Otolaryngol 2025; 145:291-298. [PMID: 39976475 DOI: 10.1080/00016489.2025.2464698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Tinnitus, a persistent condition that significantly impairs the quality of life for patients, poses a substantial burden on society. Owing to its complex and poorly understood pathogenesis, the development of effective animal models for tinnitus is imperative to enhance our comprehension of this condition. OBJECTIVES The present study aims to establish a large animal model to delineate the mechanisms underlying the onset of tinnitus. MATERIALS AND METHODS For the first time, we conducted a comparative analysis of the Post-Auricular Muscle Response (PAMR) and the Eye Blink Response (EBR) in miniature pigs, demonstrating their utility in manifesting Gap Prepulse Inhibition of the Acoustic Startle (GPIAS). Subsequently, we employed the gap-induced inhibition of PAMR to ascertain evidence of tinnitus following noise exposure. RESULTS AND CONCLUSION Through comparing the efficacy of PAMR and EBR in GPIAS detection, we have substantiated that PAMR provides a stable and objective reflection of GPIAS. Consequently, PAMR emerges as a reliable method for measuring GPIAS in miniature pigs. Furthermore, the behavioral paradigm of gap-induced inhibition of PAMR successfully identified behavioral evidence of tinnitus subsequent to noise exposure.
Collapse
Affiliation(s)
- Hanwen Zhou
- Senior Department of Otolaryngology Head and Neck Surgery, the 1th Medical Center of Chinese, PLA General Hospital, Beijing, China
- State Key Laboratory of Hearing and Balance Science, Beijing, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, China
- Ministry of Education, Key Laboratory of Hearing Science, Beijing, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Peng Liu
- Senior Department of Otolaryngology Head and Neck Surgery, the 1th Medical Center of Chinese, PLA General Hospital, Beijing, China
- State Key Laboratory of Hearing and Balance Science, Beijing, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, China
- Ministry of Education, Key Laboratory of Hearing Science, Beijing, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Chi Zhang
- Senior Department of Otolaryngology Head and Neck Surgery, the 1th Medical Center of Chinese, PLA General Hospital, Beijing, China
- State Key Laboratory of Hearing and Balance Science, Beijing, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, China
- Ministry of Education, Key Laboratory of Hearing Science, Beijing, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Xinmiao Xue
- Senior Department of Otolaryngology Head and Neck Surgery, the 1th Medical Center of Chinese, PLA General Hospital, Beijing, China
- State Key Laboratory of Hearing and Balance Science, Beijing, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, China
- Ministry of Education, Key Laboratory of Hearing Science, Beijing, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Xuemin Chen
- Senior Department of Otolaryngology Head and Neck Surgery, the 1th Medical Center of Chinese, PLA General Hospital, Beijing, China
- State Key Laboratory of Hearing and Balance Science, Beijing, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, China
- Ministry of Education, Key Laboratory of Hearing Science, Beijing, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Zhixin Zhang
- Senior Department of Otolaryngology Head and Neck Surgery, the 1th Medical Center of Chinese, PLA General Hospital, Beijing, China
- State Key Laboratory of Hearing and Balance Science, Beijing, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, China
- Ministry of Education, Key Laboratory of Hearing Science, Beijing, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Weidong Shen
- Senior Department of Otolaryngology Head and Neck Surgery, the 1th Medical Center of Chinese, PLA General Hospital, Beijing, China
- State Key Laboratory of Hearing and Balance Science, Beijing, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, China
- Ministry of Education, Key Laboratory of Hearing Science, Beijing, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Shiming Yang
- Senior Department of Otolaryngology Head and Neck Surgery, the 1th Medical Center of Chinese, PLA General Hospital, Beijing, China
- State Key Laboratory of Hearing and Balance Science, Beijing, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, China
- Ministry of Education, Key Laboratory of Hearing Science, Beijing, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Mengyun Fan
- Department of Otorhinolaryngology and Head and Neck Surgery, Xi'an Children's Hospital, Xi'an, China
| | - Fangyuan Wang
- Senior Department of Otolaryngology Head and Neck Surgery, the 1th Medical Center of Chinese, PLA General Hospital, Beijing, China
- State Key Laboratory of Hearing and Balance Science, Beijing, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, China
- Ministry of Education, Key Laboratory of Hearing Science, Beijing, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, China
| |
Collapse
|
3
|
Huang W, Cano JC, Fénelon K. Deciphering the role of brainstem glycinergic neurons during startle and prepulse inhibition. Brain Res 2024; 1836:148938. [PMID: 38615924 DOI: 10.1016/j.brainres.2024.148938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/29/2024] [Accepted: 04/11/2024] [Indexed: 04/16/2024]
Abstract
Prepulse inhibition (PPI) of the auditory startle response, a key measure of sensorimotor gating, diminishes with age and is impaired in various neurological conditions. While PPI deficits are often associated with cognitive impairments, their reversal is routinely used in experimental systems for antipsychotic drug screening. Yet, the cellular and circuit-level mechanisms of PPI remain unclear, even under non-pathological conditions. We recently showed that brainstem neurons located in the caudal pontine reticular nucleus (PnC) expressing the glycine transporter type 2 (GlyT2±) receive inputs from the central nucleus of the amygdala (CeA) and contribute to PPI but via an uncharted pathway. Here, using tract-tracing, immunohistochemistry and in vitro optogenetic manipulations coupled to field electrophysiological recordings, we reveal the neuroanatomical distribution of GlyT2± PnC neurons and PnC-projecting CeA glutamatergic neurons and we provide mechanistic insights on how these glutamatergic inputs suppress auditory neurotransmission in PnC sections. Additionally, in vivo experiments using GlyT2-Cre mice confirm that optogenetic activation of GlyT2± PnC neurons enhances PPI and is sufficient to induce PPI in young mice, emphasizing their role. However, in older mice, PPI decline is not further influenced by inhibiting GlyT2± neurons. This study highlights the importance of GlyT2± PnC neurons in PPI and underscores their diminished activity in age-related PPI decline.
Collapse
Affiliation(s)
- Wanyun Huang
- Biology Department, University of Massachusetts Amherst, Life Science Laboratories, 240 Thatcher Road, Amherst, MA, 01002, USA
| | - Jose C Cano
- Department of Biological Sciences, University of Texas at El Paso, 500 West University Avenue, El Paso, TX, 79912, USA
| | - Karine Fénelon
- Biology Department, University of Massachusetts Amherst, Life Science Laboratories, 240 Thatcher Road, Amherst, MA, 01002, USA.
| |
Collapse
|
4
|
Ortiz EA, Campbell PD, Nelson JC, Granato M. A single base pair substitution in zebrafish distinguishes between innate and acute startle behavior regulation. PLoS One 2024; 19:e0300529. [PMID: 38498506 PMCID: PMC10947677 DOI: 10.1371/journal.pone.0300529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 02/26/2024] [Indexed: 03/20/2024] Open
Abstract
Behavioral thresholds define the lowest stimulus intensities sufficient to elicit a behavioral response. Establishment of baseline behavioral thresholds during development is critical for proper responses throughout the animal's life. Despite the relevance of such innate thresholds, the molecular mechanisms critical to establishing behavioral thresholds during development are not well understood. The acoustic startle response is a conserved behavior whose threshold is established during development yet is subsequently acutely regulated. We have previously identified a zebrafish mutant line (escapist) that displays a decreased baseline or innate acoustic startle threshold. Here, we identify a single base pair substitution on Chromosome 25 located within the coding sequence of the synaptotagmin 7a (syt7a) gene that is tightly linked to the escapist acoustic hypersensitivity phenotype. By generating animals in which we deleted the syt7a open reading frame, and subsequent complementation testing with the escapist line, we demonstrate that loss of syt7a function is not the cause of the escapist behavioral phenotype. Nonetheless, escapist mutants provide a powerful tool to decipher the overlap between acute and developmental regulation of behavioral thresholds. Extensive behavioral analyses reveal that in escapist mutants the establishment of the innate acoustic startle threshold is impaired, while regulation of its acute threshold remains intact. Moreover, our behavioral analyses reveal a deficit in baseline responses to visual stimuli, but not in the acute regulation of responses to visual stimuli. Together, this work eliminates loss of syt7a as causative for the escapist phenotype and suggests that mechanisms that regulate the establishment of behavioral thresholds in escapist larvae can operate independently from those regulating acute threshold regulation.
Collapse
Affiliation(s)
- Elelbin A. Ortiz
- Department of Neuroscience, University of Pennsylvania, Pennsylvania, PA, United States of America
- Department of Cell and Developmental Biology, University of Pennsylvania, Pennsylvania, PA, United States of America
| | - Philip D. Campbell
- Department of Cell and Developmental Biology, University of Pennsylvania, Pennsylvania, PA, United States of America
- Department of Psychiatry, University of Pennsylvania, Pennsylvania, PA, United States of America
| | - Jessica C. Nelson
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Michael Granato
- Department of Cell and Developmental Biology, University of Pennsylvania, Pennsylvania, PA, United States of America
| |
Collapse
|
5
|
Luo Y, Yu Y, He H, Fan N. Acute ketamine induces neuronal hyperexcitability and deficits in prepulse inhibition by upregulating IL-6. Prog Neuropsychopharmacol Biol Psychiatry 2024; 130:110913. [PMID: 38103855 DOI: 10.1016/j.pnpbp.2023.110913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/04/2023] [Accepted: 12/10/2023] [Indexed: 12/19/2023]
Abstract
Acute ketamine administration results in psychotic symptoms similar to those observed in schizophrenia and is regarded as a pharmacological model of schizophrenia. Accumulating evidence suggests that patients with schizophrenia show increased IL-6 levels in the blood and cerebrospinal fluid and that IL-6 levels are associated with the severity of psychotic symptoms. In the present study, we found that a single ketamine exposure led to increased expression of IL-6 and IL-6Rα, decreased dendritic spine density, increased expression and currents of T-type calcium channels, and increased neuron excitability in the hippocampal CA1 area 12 h after exposure. Acute ketamine administration also led to impaired prepulse inhibition (PPI) 12 h after administration. Additionally, we found that the expression of signaling molecules IKKα/β, NF-κB, JAK2, and STAT3 was upregulated 12 h after a single ketamine injection. The decreases in dendritic spine density, the increases in calcium currents and neuron excitability, and the impairments in PPI were ameliorated by blocking IL-6 or IL-6Rα. Our findings show that blocking IL-6 or its receptor may protect hippocampal neurons from hyperexcitability, thereby ameliorating ketamine-induced psychotic effects. Our study provides additional evidence that targeting IL-6 and its receptor is a potential strategy for treating psychotic symptoms in acute ketamine-induced psychosis and schizophrenia.
Collapse
Affiliation(s)
- Yayan Luo
- The Affiliated Brain Hospital of Guangzhou Medical University, 36 Mingxin Road, Liwan District, Guangzhou, Guangdong Province 510370, China
| | - Yang Yu
- The Affiliated Brain Hospital of Guangzhou Medical University, 36 Mingxin Road, Liwan District, Guangzhou, Guangdong Province 510370, China
| | - Hongbo He
- The Affiliated Brain Hospital of Guangzhou Medical University, 36 Mingxin Road, Liwan District, Guangzhou, Guangdong Province 510370, China
| | - Ni Fan
- The Affiliated Brain Hospital of Guangzhou Medical University, 36 Mingxin Road, Liwan District, Guangzhou, Guangdong Province 510370, China.
| |
Collapse
|
6
|
Read E, Hindges R. A novel locomotion-based prepulse inhibition assay in zebrafish larvae. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.000914. [PMID: 38344062 PMCID: PMC10853821 DOI: 10.17912/micropub.biology.000914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 12/20/2023] [Accepted: 01/11/2024] [Indexed: 03/07/2024]
Abstract
Sensory gating, measured using prepulse inhibition (PPI), is an endophenotype of neuropsychiatric disorders that can be assessed in larval zebrafish models. However, current PPI assays require high-speed cameras to capture rapid c-bend startle behaviours of the larvae. In this study, we designed and employed a PPI paradigm that uses locomotion as a read-out of zebrafish larval startle responses. PPI percentage was measured at a maximum of 87% and strongly reduced upon administration of the NMDA receptor antagonist, MK-801. This work provides the foundation for simpler and more accessible PPI assays using larval zebrafish to model key endophenotypes of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Emily Read
- Centre for Developmental Neurobiology & MRC Centre for Neurodevelopmental Disorders, King's College London, London, England, United Kingdom
| | - Robert Hindges
- Centre for Developmental Neurobiology & MRC Centre for Neurodevelopmental Disorders, King's College London, London, England, United Kingdom
| |
Collapse
|
7
|
El-Cheikh Mohamad A, Möhrle D, Haddad FL, Rose A, Allman BL, Schmid S. Assessing the Cntnap2 knockout rat prepulse inhibition deficit through prepulse scaling of the baseline startle response curve. Transl Psychiatry 2023; 13:321. [PMID: 37852987 PMCID: PMC10584930 DOI: 10.1038/s41398-023-02629-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/27/2023] [Accepted: 10/06/2023] [Indexed: 10/20/2023] Open
Abstract
Many neurodevelopmental disorders, including autism spectrum disorder (ASD), are associated with changes in sensory processing and sensorimotor gating. The acoustic startle response and prepulse inhibition (PPI) of startle are widely used translational measures for assessing sensory processing and sensorimotor gating, respectively. The Cntnap2 knockout (KO) rat has proven to be a valid model for ASD, displaying core symptoms, including sensory processing perturbations. Here, we used a novel method to assess startle and PPI in Cntnap2 KO rats that allows for the identification of separate scaling components: startle scaling, which is a change in startle amplitude to a given sound, and sound scaling, which reflects a change in sound processing. Cntnap2 KO rats show increased startle due to both an increased overall response (startle scaling) and a left shift of the sound/response curve (sound scaling). In the presence of a prepulse, wildtype rats show a reduction of startle due to both startle scaling and sound scaling, whereas Cntnap2 KO rats show normal startle scaling, but disrupted sound scaling, resulting in the reported PPI deficit. These results validate that startle and sound scaling by a prepulse are indeed two independent processes, with only the latter being impaired in Cntnap2 KO rats. As startle scaling is likely related to motor output and sound scaling to sound processing, this novel approach reveals additional information on the possible cause of PPI disruptions in preclinical models.
Collapse
Affiliation(s)
- Alaa El-Cheikh Mohamad
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Dorit Möhrle
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Faraj L Haddad
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Anton Rose
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Brian L Allman
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Susanne Schmid
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada.
- Department of Psychology, University of Western Ontario, London, ON, Canada.
| |
Collapse
|
8
|
Han S, Zhuang D, Wang J, Ju C. Inhibition of neuronal Kv7 channels ameliorates MK-801-induced cognitive dysfunction in mice via up-regulating NAMPT expression. Neurosci Lett 2023; 814:137471. [PMID: 37673371 DOI: 10.1016/j.neulet.2023.137471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/01/2023] [Accepted: 09/04/2023] [Indexed: 09/08/2023]
Abstract
PURPOSE Abnormal energy metabolism affects cognitive function in schizophrenia. Nicotinamide phosphoribosyltransferase (NAMPT), as the rate-limiting enzyme of nicotinamide adenine dinucleotide (NAD+), is involved in energy metabolism by regulating the synthesis of NAD+. This study aims to clarify whether inhibition of Kv7 channels improves cognitive impairment by up-regulating NAMPT expression to increase the level of NAD+. METHODS The dominant negative pore mutation of KCNQ2 in transgenic mice was achieved by mutating residual 279-Gly to Ser (rQ2-G279S). A cognitive deficit model was established by injecting MK-801 into C57BL/6J mice. Y-maze and prepulse inhibition (PPI) tests were performed to evaluate cognitive ability. Gene and protein expression of NAMPT in the mouse hippocampus, cortex, and PC-12 cells were measured by qRT-PCR and Western blot. The level of NAD+ was measured by a WST-8 assay. RESULTS The Y-maze and PPI results showed that genetic or pharmacological inhibition of Kv7 channels by XE991 enhanced cognitive function in mice. Furthermore, inhibition of Kv7 channels increased the gene and protein expression of NAMPT and the level of NAD+ in the hippocampus and cortex of the above animal model. Similarly, XE991 treatment increased NAMPT expression and NAD+ levels in PC-12 cells. NAMPT inhibitor FK866 and Kv7 channel opener retigabine reversed the effects of XE991 in vivo and in vitro. In addition, XE991 increased pAMPK protein expression in PC-12 cells, while AMPK inhibitor Compound C counteracted the effect of XE991 on increasing NAMPT expression and NAD+ levels. CONCLUSIONS Suppression of Kv7 channel function improved spatial working memory and PPI impairment. This result may be achieved by activating AMPK to up-regulate NAMPT expression and thus increase NAD+ levels.
Collapse
Affiliation(s)
- Shuo Han
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, China
| | - Dongpei Zhuang
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, China
| | - Jing Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, China; Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining 272000, China
| | - Chuanxia Ju
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, China.
| |
Collapse
|
9
|
Ortiz EA, Campbell PD, Nelson JC, Granato M. A single base pair substitution on Chromosome 25 in zebrafish distinguishes between development and acute regulation of behavioral thresholds. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.25.554673. [PMID: 37662318 PMCID: PMC10473726 DOI: 10.1101/2023.08.25.554673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Behavioral thresholds define the lowest stimulus intensities sufficient to elicit a behavioral response. Establishment of baseline behavioral thresholds during development is critical for proper responses throughout the animal's life. Despite the relevance of such innate thresholds, the molecular mechanisms critical to establishing behavioral thresholds during development are not well understood. The acoustic startle response is a conserved behavior whose threshold is established during development yet is subsequently acutely regulated. We have previously identified a zebrafish mutant line ( escapist ) that displays a decreased baseline or innate acoustic startle threshold. Here, we identify a single base pair substitution on Chromosome 25 located within the coding sequence of the synaptotagmin 7a ( syt7a ) gene that is tightly linked to the escapist acoustic hypersensitivity phenotype. By generating animals in which we deleted the syt7a open reading frame, and subsequent complementation testing with the escapist line, we demonstrate that loss of syt7a function is not the cause of the escapist behavioral phenotype. Nonetheless, escapist mutants provide a powerful tool to decipher the overlap between acute and developmental regulation of behavioral thresholds. Extensive behavioral analyses reveal that in escapist mutants the establishment of the innate acoustic startle threshold is impaired, while regulation of its acute threshold remains intact. Moreover, our behavioral analyses reveal a deficit in baseline responses to visual stimuli, but not in the acute regulation of responses to visual stimuli. Together, this work eliminates loss of syt7a as causative for the escapist phenotype and suggests that mechanisms that regulate the establishment of behavioral thresholds in escapist larvae can operate largely independently from those regulating acute threshold regulation.
Collapse
|
10
|
Schulz SE, Luszawski M, Hannah KE, Stevenson RA. Sensory Gating in Neurodevelopmental Disorders: A Scoping Review. Res Child Adolesc Psychopathol 2023; 51:1005-1019. [PMID: 37014483 DOI: 10.1007/s10802-023-01058-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2023] [Indexed: 04/05/2023]
Abstract
This review aimed to explore the current understanding of sensory gating in neurodevelopmental disorders as a possible transdiagnostic mechanism. We applied methods according to the Joanna Briggs Institute Manual for Evidence Synthesis, following the population, concept, and context scoping review eligibility criteria. Using a comprehensive search strategy in five relevant research databases (Medline, EMBASE, CINAHL, PsychInfo, and Scopus), we searched for relevant peer-reviewed, primary research articles and unpublished data. Two independent reviewers screened the titles and abstracts, full-texts, and completed data extraction. We identified a total of 81 relevant articles and used descriptive analyses to summarize the characteristics and outcomes of all identified studies. Literature regarding sensory gating was most common in autistic populations with relatively fewer studies examining attention-deficit/hyperactivity disorder, tic disorders, and childhood-onset fluency disorder (COFD). The methods to assess sensory gating varied widely both within and between groups and included measures such as habituation, prepulse inhibition, affect-modulated inhibition, medication and other intervention trials. Most consistently, when participants complete questionnaires about their sensory experiences, those who have neurodevelopmental disorders report differences in their sensory gating. Affect-modulated inhibition appears to be discrepant between samples with and without neurodevelopmental disorder diagnoses. Habituation was the most commonly reported phenomenon and many differences in habituation have been found in autistic individuals and individuals with tic disorders whereas concerns with inhibition seemed more common in COFD. Overall, the evidence is inconsistent within and between disorders suggesting there is still much to learn about sensory gating in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Samantha E Schulz
- Department of Psychology, University of Western Ontario, London, Canada
- Brain and Mind Institute, University of Western Ontario, London, Canada
- Western Institute for Neuroscience, University of Western Ontario, London, ON, Canada
| | - Michelle Luszawski
- Department of Psychology, University of Western Ontario, London, Canada
- Brain and Mind Institute, University of Western Ontario, London, Canada
- Western Institute for Neuroscience, University of Western Ontario, London, ON, Canada
| | - Kara E Hannah
- Department of Psychology, University of Western Ontario, London, Canada
- Brain and Mind Institute, University of Western Ontario, London, Canada
- Western Institute for Neuroscience, University of Western Ontario, London, ON, Canada
| | - Ryan A Stevenson
- Department of Psychology, University of Western Ontario, London, Canada.
- Brain and Mind Institute, University of Western Ontario, London, Canada.
- Western Institute for Neuroscience, University of Western Ontario, London, ON, Canada.
| |
Collapse
|
11
|
Zheng A, Schmid S. A review of the neural basis underlying the acoustic startle response with a focus on recent developments in mammals. Neurosci Biobehav Rev 2023; 148:105129. [PMID: 36914078 DOI: 10.1016/j.neubiorev.2023.105129] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/13/2023]
Abstract
The startle response consists of whole-body muscle contractions, eye-blink, accelerated heart rate, and freezing in response to a strong, sudden stimulus. It is evolutionarily preserved and can be observed in any animal that can perceive sensory signals, indicating the important protective function of startle. Startle response measurements and its alterations have become a valuable tool for exploring sensorimotor processes and sensory gating, especially in the context of pathologies of psychiatric disorders. The last reviews on the neural substrates underlying acoustic startle were published around 20 years ago. Advancements in methods and techniques have since allowed new insights into acoustic startle mechanisms. This review is focused on the neural circuitry that drives the primary acoustic startle response in mammals. However, there have also been very successful efforts to identify the acoustic startle pathway in other vertebrates and invertebrates in the past decades, so at the end we briefly summarize these studies and comment on the similarities and differences between species.
Collapse
Affiliation(s)
- Alice Zheng
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, Canada
| | - Susanne Schmid
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, Canada.
| |
Collapse
|
12
|
Unraveling Presenilin 2 Functions in a Knockout Zebrafish Line to Shed Light into Alzheimer's Disease Pathogenesis. Cells 2023; 12:cells12030376. [PMID: 36766721 PMCID: PMC9913325 DOI: 10.3390/cells12030376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023] Open
Abstract
Mutations in presenilin 2 (PS2) have been causally linked to the development of inherited Alzheimer's disease (AD). Besides its role as part of the γ-secretase complex, mammalian PS2 is also involved, as an individual protein, in a growing number of cell processes, which result altered in AD. To gain more insight into PS2 (dys)functions, we have generated a presenilin2 (psen2) knockout zebrafish line. We found that the absence of the protein does not markedly influence Notch signaling at early developmental stages, suggesting a Psen2 dispensable role in the γ-secretase-mediated Notch processing. Instead, loss of Psen2 induces an exaggerated locomotor response to stimulation in fish larvae, a reduced number of ER-mitochondria contacts in zebrafish neurons, and an increased basal autophagy. Moreover, the protein is involved in mitochondrial axonal transport, since its acute downregulation reduces in vivo organelle flux in zebrafish sensory neurons. Importantly, the expression of a human AD-linked mutant of the protein increases this vital process. Overall, our results confirm zebrafish as a good model organism for investigating PS2 functions in vivo, representing an alternative tool for the characterization of new AD-linked defective cell pathways and the testing of possible correcting drugs.
Collapse
|
13
|
Volnova A, Kurzina N, Belskaya A, Gromova A, Pelevin A, Ptukha M, Fesenko Z, Ignashchenkova A, Gainetdinov RR. Noradrenergic Modulation of Learned and Innate Behaviors in Dopamine Transporter Knockout Rats by Guanfacine. Biomedicines 2023; 11:222. [PMID: 36672730 PMCID: PMC9856099 DOI: 10.3390/biomedicines11010222] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Investigation of the precise mechanisms of attention deficit and hyperactivity disorder (ADHD) and other dopamine-associated conditions is crucial for the development of new treatment approaches. In this study, we assessed the effects of repeated and acute administration of α2A-adrenoceptor agonist guanfacine on innate and learned forms of behavior of dopamine transporter knockout (DAT-KO) rats to evaluate the possible noradrenergic modulation of behavioral deficits. DAT-KO and wild type rats were trained in the Hebb-Williams maze to perform spatial working memory tasks. Innate behavior was evaluated via pre pulse inhibition (PPI). Brain activity of the prefrontal cortex and the striatum was assessed. Repeated administration of GF improved the spatial working memory task fulfillment and PPI in DAT-KO rats, and led to specific changes in the power spectra and coherence of brain activity. Our data indicate that both repeated and acute treatment with a non-stimulant noradrenergic drug lead to improvements in the behavior of DAT-KO rats. This study further supports the role of the intricate balance of norepinephrine and dopamine in the regulation of attention. The observed compensatory effect of guanfacine on the behavior of hyperdopaminergic rats may be used in the development of combined treatments to support the dopamine-norepinephrine balance.
Collapse
Affiliation(s)
- Anna Volnova
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg 199034, Russia
- Biological Faculty, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | - Natalia Kurzina
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | - Anastasia Belskaya
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | - Arina Gromova
- Biological Faculty, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | - Arseniy Pelevin
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg 199034, Russia
- Biological Faculty, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | - Maria Ptukha
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | - Zoia Fesenko
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | | | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg 199034, Russia
- Saint Petersburg University Hospital, Saint Petersburg 199034, Russia
| |
Collapse
|
14
|
Carroll JB, Hamidi S, Gabriele ML. Microglial heterogeneity and complement component 3 elimination within emerging multisensory midbrain compartments during an early critical period. Front Neurosci 2023; 16:1072667. [PMID: 36685243 PMCID: PMC9846048 DOI: 10.3389/fnins.2022.1072667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/09/2022] [Indexed: 01/05/2023] Open
Abstract
The lateral cortex of the inferior colliculus (LCIC) is a midbrain shell region that receives multimodal inputs that target discrete zones of its compartmental (modular-matrix) framework. This arrangement emerges perinatally in mice (postnatal day, P0-P12) as somatosensory and auditory inputs segregate into their respective modular and matrix terminal patterns. Microglial cells (MGCs) perform a variety of critical functions in the developing brain, among them identifying areas of active circuit assembly and selectively pruning exuberant or underutilized connections. Recent evidence in other brain structures suggest considerable MGC heterogeneity across the lifespan, particularly during established developmental critical periods. The present study examines the potential involvement of classical complement cascade signaling (C3-CR3/CD11b) in refining early multisensory networks, and identifies several microglial subsets exhibiting distinct molecular signatures within the nascent LCIC. Immunostaining was performed in GAD67-green fluorescent protein (GFP) and CX3CR1-GFP mice throughout and after the defined LCIC critical period. GAD labeling highlights the emerging LCIC modularity, while CX3CR1 labeling depicts MGCs expressing the fractalkine receptor. C3 expression is widespread throughout the LCIC neuropil early on, prior to its conspicuous absence from modular zones at P8, and more global disappearance following critical period closure. CD11b-expressing microglia while homogeneously distributed at birth, are biased to modular fields at P8 and then the surrounding matrix by P12. Temporal and spatial matching of the disappearance of C3 by LCIC compartment (i.e., modules then matrix) with CD11b-positive MGC occupancy implicates complement signaling in the selective refinement of early LCIC connectivity. Multiple-labeling studies for a variety of established MGC markers (CD11b, CX3CR1, Iba1, TMEM119) indicate significant MGC heterogeneity in the LCIC as its compartments and segregated multisensory maps emerge. Marker colocalization was the exception rather than the rule, suggesting that unique MGC subpopulations exist in the LCIC and perhaps serve distinct developmental roles. Potential mechanisms whereby microglia sculpt early multisensory LCIC maps and how such activity/inactivity may underlie certain neurodevelopmental conditions, including autism spectrum disorder and schizophrenia, are discussed.
Collapse
Affiliation(s)
| | | | - Mark L. Gabriele
- Department of Biology, James Madison University, Harrisonburg, VA, United States
| |
Collapse
|
15
|
Zlomuzica A, Plank L, Dere E. A new path to mental disorders: Through gap junction channels and hemichannels. Neurosci Biobehav Rev 2022; 142:104877. [PMID: 36116574 DOI: 10.1016/j.neubiorev.2022.104877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 08/20/2022] [Accepted: 09/13/2022] [Indexed: 11/18/2022]
Abstract
Behavioral disturbances related to emotional regulation, reward processing, cognition, sleep-wake regulation and activity/movement represent core symptoms of most common mental disorders. Increasing empirical and theoretical evidence suggests that normal functioning of these behavioral domains relies on fine graded coordination of neural and glial networks which are maintained and modulated by intercellular gap junction channels and unapposed pannexin or connexin hemichannels. Dysfunctions in these networks might contribute to the development and maintenance of psychopathological and neurobiological features associated with mental disorders. Here we review and discuss the evidence indicating a prominent role of gap junction channel and hemichannel dysfunction in core symptoms of mental disorders. We further discuss how the increasing knowledge on intercellular gap junction channels and unapposed pannexin or connexin hemichannels in the brain might lead to deeper mechanistic insight in common mental disorders and to the development of novel treatment approaches. We further attempt to exemplify what type of future research on this topic could be integrated into multidimensional approaches to understand and cure mental disorders.
Collapse
Affiliation(s)
- Armin Zlomuzica
- Department of Behavioral and Clinical Neuroscience, Ruhr-University Bochum (RUB), Massenbergstraße 9-13, D-44787 Bochum, Germany.
| | - Laurin Plank
- Department of Behavioral and Clinical Neuroscience, Ruhr-University Bochum (RUB), Massenbergstraße 9-13, D-44787 Bochum, Germany
| | - Ekrem Dere
- Department of Behavioral and Clinical Neuroscience, Ruhr-University Bochum (RUB), Massenbergstraße 9-13, D-44787 Bochum, Germany; Sorbonne Université. Institut de Biologie Paris-Seine, (IBPS), Département UMR 8256: Adaptation Biologique et Vieillissement, UFR des Sciences de la Vie, Campus Pierre et Marie Curie, Bâtiment B, 9 quai Saint Bernard, F-75005 Paris, France.
| |
Collapse
|
16
|
Beppi C, Penner M, Straumann D, Bögli SY. A non-invasive biomechanical model of mild TBI in larval zebrafish. PLoS One 2022; 17:e0268901. [PMID: 35622781 PMCID: PMC9140253 DOI: 10.1371/journal.pone.0268901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 05/05/2022] [Indexed: 11/18/2022] Open
Abstract
A mild traumatic brain injury is a neurological dysfunction caused by biomechanical forces transmitted to the brain in physical impacts. The current understanding of the neuropathological cascade resulting in the manifested clinical signs and symptoms is limited due to the absence of sensitive brain imaging methods. Zebrafish are established models for the reproduction and study of neurobiological pathologies. However, all available models mostly recreate moderate-to-severe focal injuries in adult zebrafish. The present work has induced a mild brain trauma in larval zebrafish through a non-invasive biomechanical approach. A custom-made apparatus with a commercially available motor was employed to expose larvae to rapidly decelerating linear movements. The neurophysiological changes following concussion were assessed through behavioural quantifications of startle reflex locomotor distance and habituation metrics. Here we show that the injury was followed, within five minutes, by a transient anxiety state and CNS dysfunction manifested by increased startle responsivity with impaired startle habituation, putatively mirroring the human clinical sign of hypersensitivity to noise. Within a day after the injury, chronic effects arose, as evidenced by an overall reduced responsivity to sensory stimulation (lower amplitude and distance travelled along successive stimuli), reflecting the human post-concussive symptomatology. This study represents a step forward towards the establishment of a parsimonious (simple, less ethically concerning, yet sensitive) animal model of mild TBI. Our behavioural findings mimic aspects of acute and chronic effects of human concussion, which warrant further study at molecular, cellular and circuit levels. While our model opens wide avenues for studying the underlying cellular and molecular pathomechanisms, it also enables high-throughput testing of therapeutic interventions to accelerate post-concussive recovery.
Collapse
Affiliation(s)
- Carolina Beppi
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Swiss Concussion Center, Schulthess Clinic, Zurich, Switzerland
- * E-mail:
| | - Marco Penner
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Dominik Straumann
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Swiss Concussion Center, Schulthess Clinic, Zurich, Switzerland
| | - Stefan Yu Bögli
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Swiss Concussion Center, Schulthess Clinic, Zurich, Switzerland
| |
Collapse
|
17
|
Li W, Mao Z, Bo Q, Sun Y, Wang Z, Wang C. Pre-pulse inhibition deficits in individuals at clinical high-risk for psychosis: A systematic review and meta-analysis. Early Interv Psychiatry 2021; 15:794-806. [PMID: 32705810 DOI: 10.1111/eip.13015] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 06/15/2020] [Accepted: 06/24/2020] [Indexed: 11/29/2022]
Abstract
AIM Neurophysiological markers of schizophrenia may help identify individuals who are at an increased risk of developing psychosis. As an operational measure of sensorimotor gating, pre-pulse inhibition (PPI) deficit has been investigated in clinical high-risk (CHR) individuals. In this study, we performed a systematic review and meta-analysis of studies that investigated PPI in CHR individuals. METHODS Relevant studies published as of July 2019 were retrieved from the PubMed, Cochrane, Embase, PscyINFO, EBSCO and Chinese databases. PPI was evaluated by calculating the standard mean differences (SMDs) between CHR individuals and healthy controls (HC) in meta-analysis. Quality of studies was assessed using the Newcastle-Ottawa Scale. I2 index was used to assess heterogeneity and Egger's test was used to assess publication bias. RESULTS Eight studies were found to be eligible. The meta-analysis included five studies with a combined study population of 184 CHR subjects and 161 HC. CHR individuals showed lower PPI levels compared to HC in 120 ms inter-stimulus interval or stimulus onset asynchrony paradigm (P = .491, SMD = -0.62). No significant heterogeneity was observed in 120 ms PPI paradigm (χ2 = 3.41, P = .491, I2 = 0.0%). CONCLUSION CHR individuals had lower PPI level compared to HC in 120 ms paradigm, which were relatively stable and significant. The results indicate the presence of information processing and inhibitory problems prior to the development of full-blown psychosis. PPI may be clinically used as an objective indicator to supplement the understanding of CHR individuals.
Collapse
Affiliation(s)
- Weidi Li
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders & Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Zhen Mao
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders & Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Qijing Bo
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders & Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yue Sun
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders & Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Zhimin Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders & Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Chuanyue Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders & Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| |
Collapse
|
18
|
Li W, Mao Z, Bo Q, Sun Y, Wang Z, Wang C. Prepulse inhibition in first-degree relatives of schizophrenia patients: A systematic review. Early Interv Psychiatry 2021; 15:652-661. [PMID: 32567764 DOI: 10.1111/eip.13003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/15/2020] [Accepted: 05/24/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Prepulse inhibition (PPI) is a measure of sensorimotor gating used to identify deficits in early-stage information processing and inhibitory function defects. Many studies support the presence of PPI deficits in schizophrenia patients. However, very few studies have explored PPI levels among first-degree relatives (FDR) of schizophrenia patients, and the results have been inconsistent. This review article explored PPI levels in FDR of schizophrenia patients. METHODS We performed a systematic literature review using the PubMed, Cochrane, Embase, EBSCO and Chinese databases from inception to January 2020. A series of related factors (eg, PPI paradigm, heritability and sample characteristics) and outcomes were summarized from the literature that met the inclusion criteria. The Newcastle-Ottawa Scale was used to assess the quality of the included studies. RESULTS A total of eight studies were eligible for systematic review after screening. A meta-analysis of the selected studies was not conducted due to the limitations of quantity and paradigm heterogeneity. A majority of the studies' subjects were siblings of schizophrenia patients and different paradigms were applied. Most of the included studies reported no difference in PPI values between FDR of schizophrenia patients and healthy controls. CONCLUSION Contrary to traditional certainty that unaffected FDR of schizophrenia patients have PPI defects, our review found no sufficient evidence supporting that the PPI level in FDR of schizophrenia patients was lower than in healthy controls. A prospective cohort study focusing on different outcomes such as developing schizophrenia is required to explore PPI levels in FDR of schizophrenia patients.
Collapse
Affiliation(s)
- Weidi Li
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders & Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Zhen Mao
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders & Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Qijing Bo
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders & Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yue Sun
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders & Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Zhimin Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders & Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Chuanyue Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders & Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
A model-based quantification of startle reflex habituation in larval zebrafish. Sci Rep 2021; 11:846. [PMID: 33436805 PMCID: PMC7804396 DOI: 10.1038/s41598-020-79923-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 12/14/2020] [Indexed: 01/29/2023] Open
Abstract
Zebrafish is an established animal model for the reproduction and study of neurobiological pathogenesis of human neurological conditions. The 'startle reflex' in zebrafish larvae is an evolutionarily preserved defence response, manifesting as a quick body-bend in reaction to sudden sensory stimuli. Changes in startle reflex habituation characterise several neuropsychiatric disorders and hence represent an informative index of neurophysiological health. This study aimed at establishing a simple and reliable experimental protocol for the quantification of startle reflex response and habituation. The fish were stimulated with 20 repeated pulses of specific vibratory frequency, acoustic intensity/power, light-intensity and interstimulus-interval, in three separate studies. The cumulative distance travelled, namely the sum of the distance travelled (mm) during all 20 stimuli, was computed as a group-level description for all the experimental conditions in each study. Additionally, by the use of bootstrapping, the data was fitted to a model of habituation with a first-order exponential representing the decay of locomotor distance travelled over repeated stimulation. Our results suggest that startle habituation is a stereotypic first-order process with a decay constant ranging from 1 to 2 stimuli. Habituation memory lasts no more than 5 min, as manifested by the locomotor activity recovering to baseline levels. We further observed significant effects of vibratory frequency, acoustic intensity/power and interstimulus-interval on the amplitude, offset, decay constant and cumulative distance travelled. Instead, the intensity of the flashed light did not contribute to significant behavioural variations. The findings provide novel insights as to the influence of different stimuli parameters on the startle reflex habituation and constitute a helpful reference framework for further investigation.
Collapse
|
20
|
ZFP804A mutant mice display sex-dependent schizophrenia-like behaviors. Mol Psychiatry 2021; 26:2514-2532. [PMID: 33303946 PMCID: PMC8440220 DOI: 10.1038/s41380-020-00972-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 10/20/2020] [Accepted: 11/24/2020] [Indexed: 12/22/2022]
Abstract
Genome-wide association studies uncovered the association of ZNF804A (Zinc-finger protein 804A) with schizophrenia (SZ). In vitro data have indicated that ZNF804A might exert its biological roles by regulating spine and neurite morphogenesis. However, no in vivo data are available for the role of ZNF804A in psychiatric disorders in general, SZ in particular. We generated ZFP804A mutant mice, and they showed deficits in contextual fear and spatial memory. We also observed the sensorimotor gating impairment, as revealed by the prepulse inhibition test, but only in female ZFP804A mutant mice from the age of 6 months. Notably, the PPI difference between the female mutant and control mice was no longer existed with the administration of Clozapine or after the ovariectomy. Hippocampal long-term potentiation was normal in both genders of the mutant mice. Long-term depression was absent in male mutants, but facilitated in the female mutants. Protein levels of hippocampal serotonin-6 receptor and GABAB1 receptor were increased, while those of cortical dopamine 2 receptor were decreased in the female mutants with no obvious changes in the male mutants. Moreover, the spine density was reduced in the cerebral cortex and hippocampus of the mutant mice. Knockdown of ZFP804A impaired the neurite morphogenesis of cortical and hippocampal neurons, while its overexpression enhanced neurite morphogenesis only in the cortical neurons in vitro. Our data collectively support the idea that ZFP804A/ZNF804A plays important roles in the cognitive functions and sensorimotor gating, and its dysfunction may contribute to SZ, particularly in the female patients.
Collapse
|
21
|
Péczely L, Kékesi G, Kállai V, Ollmann T, László K, Büki A, Lénárd L, Horváth G. Effects of D 2 dopamine receptor activation in the ventral pallidum on sensory gating and food-motivated learning in control and schizophrenia model (Wisket) rats. Behav Brain Res 2020; 400:113047. [PMID: 33279633 DOI: 10.1016/j.bbr.2020.113047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 11/28/2022]
Abstract
Dopamine D2 receptors (D2Rs) of the ventral pallidum (VP) play important role in motivational and learning processes, however, their potential role in triggering schizophrenic symptoms has not been investigated, yet. In the present experiments the effects of locally administered D2R agonist quinpirole were investigated on behavioral parameters related to sensorimotor gating, motor activity and food-motivated labyrinth learning. Two weeks after bilateral implantation of microcannulae into the VP, the acute (30 min) and delayed (3, 21 and 24 h) effects of quinpirole microinjection (1 μg/0.4 μL at both sides) were investigated in Wistar and schizophrenia model (Wisket substrain) rats in prepulse inhibition (PPI) and the reward-based Ambitus tests. Quinpirole administration did not modify the impaired sensorimotor gating in Wisket rats, but it led to significant deficit in Wistar animals. Regarding the locomotor activity in the Ambitus test, no effects of quinpirole were detected in either groups at the investigated time points. In contrast, quinpirole resulted in decreased exploratory and food-collecting activities in Wistar rats with 21 and 24 h delay. Though, impaired food-related motivation could be observed in Wisket rats, but quinpirole treatment did not result in further deterioration. In summary, our results showed that the VP D2R activation in Wistar rats induces symptoms similar to those observed in schizophrenia model Wisket rats. These data suggest that Wisket rats might have significant alterations in the functional activity of VP, which might be due to its enhanced dopaminergic activity.
Collapse
Affiliation(s)
- László Péczely
- Institute of Physiology, Faculty of Medicine, University of Pécs, Pécs, Hungary; Centre for Neuroscience, Pécs University, Pécs, Hungary.
| | - Gabriella Kékesi
- Department of Physiology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Veronika Kállai
- Institute of Physiology, Faculty of Medicine, University of Pécs, Pécs, Hungary; Centre for Neuroscience, Pécs University, Pécs, Hungary
| | - Tamás Ollmann
- Institute of Physiology, Faculty of Medicine, University of Pécs, Pécs, Hungary; Centre for Neuroscience, Pécs University, Pécs, Hungary
| | - Kristóf László
- Institute of Physiology, Faculty of Medicine, University of Pécs, Pécs, Hungary; Centre for Neuroscience, Pécs University, Pécs, Hungary
| | - Alexandra Büki
- Department of Physiology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - László Lénárd
- Institute of Physiology, Faculty of Medicine, University of Pécs, Pécs, Hungary; Molecular Neuroendocrinology and Neurophysiology Research Group, Szentágothai Research Centre, Pécs University, Pécs, Hungary; Centre for Neuroscience, Pécs University, Pécs, Hungary
| | - Gyöngyi Horváth
- Department of Physiology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
22
|
Sex-Specific Role for Dopamine Receptor D2 in Dorsal Raphe Serotonergic Neuron Modulation of Defensive Acoustic Startle and Dominance Behavior. eNeuro 2020; 7:ENEURO.0202-20.2020. [PMID: 33214315 PMCID: PMC7768286 DOI: 10.1523/eneuro.0202-20.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 10/19/2020] [Accepted: 11/09/2020] [Indexed: 11/27/2022] Open
Abstract
Brain networks underlying states of social and sensory alertness are normally adaptive, influenced by serotonin and dopamine (DA), and abnormal in neuropsychiatric disorders, often with sex-specific manifestations. Underlying circuits, cells, and molecules are just beginning to be delineated. Implicated is a subtype of serotonergic neuron denoted Drd2-Pet1, distinguished by expression of the type-2 DA receptor (Drd2) gene, inhibited cell-autonomously by DRD2 agonism in slice, and, when constitutively silenced in male mice, affects levels of defensive and exploratory behaviors (Niederkofler et al., 2016). Unknown has been whether DRD2 signaling in these Pet1 neurons contributes to their capacity for shaping defensive behaviors. To address this, we generated mice in which Drd2 gene sequences were deleted selectively in Pet1 neurons. We found that Drd2Pet1-CKO males, but not females, demonstrated increased winning against sex-matched controls in a social dominance assay. Drd2Pet1-CKO females, but not males, exhibited blunting of the acoustic startle response, a protective, defensive reflex. Indistinguishable from controls were auditory brainstem responses (ABRs), locomotion, cognition, and anxiety-like and depression-like behaviors. Analyzing wild-type Drd2-Pet1 neurons, we found sex-specific differences in the proportional distribution of axonal collaterals, in action potential (AP) duration, and in transcript levels of Gad2, important for GABA synthesis. Drd2Pet1-CKO cells displayed sex-specific differences in the percentage of cells harboring Gad2 transcripts. Our results suggest that DRD2 function in Drd2-Pet1 neurons is required for normal defensive/protective behaviors in a sex-specific manner, which may be influenced by the identified sex-specific molecular and cellular features. Related behaviors in humans too show sex differences, suggesting translational relevance.
Collapse
|
23
|
Wang J, Yu W, Gao Q, Ju C, Wang K. Prefrontal inhibition of neuronal K v 7 channels enhances prepulse inhibition of acoustic startle reflex and resistance to hypofrontality. Br J Pharmacol 2020; 177:4720-4733. [PMID: 32839968 PMCID: PMC7520443 DOI: 10.1111/bph.15236] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/31/2020] [Accepted: 08/06/2020] [Indexed: 11/28/2022] Open
Abstract
Background and Purpose Dysfunction of the prefrontal cortex (PFC) is involved in the cognitive deficits in neuropsychiatric diseases, such as schizophrenia, characterized by deficient neurotransmission known as NMDA receptor hypofrontality. Thus, enhancing prefrontal activity may alleviate hypofrontality‐induced cognitive deficits. To test this hypothesis, we investigated the effect of forebrain‐specific suppression or pharmacological inhibition of native Kv7/KCNQ/M‐current on glutamatergic hypofrontality induced by the NMDA receptor antagonist MK‐801. Experimental Approach The forebrain‐specific inhibition of native M‐current was generated by transgenic expression, in mice, of a dominant‐negative pore mutant G279S of Kv7.2/KCNQ2 channels that suppresses channel function. A mouse model of cognitive impairment was established by single i.p. injection of 0.1 mg·kg−1 MK‐801. Mouse models of prepulse inhibition (PPI) of acoustic startle reflex and Y‐maze spontaneous alternation test were used for evaluation of cognitive behaviour. Hippocampal brain slice recordings of LTP were used to assess synaptic plasticity. Hippocampus and cortex were dissected for detecting protein expression using western blot analysis. Key Results Genetic suppression of Kv7 channel function in the forebrain or pharmacological inhibition of Kv7 channels by the specific blocker XE991 enhanced PPI and also alleviated MK‐801 induced cognitive decline. XE991 also attenuated MK‐801‐induced LTP deficits and increased basal synaptic transmissions. Western blot analysis revealed that inhibiting Kv7 channels resulted in elevation of pAkt1 and pGSK‐3β expressions in both hippocampus and cortex. Conclusions and Implications Both genetic and pharmacological inhibition of Kv7 channels alleviated PPI and cognitive deficits. Mechanistically, inhibition of Kv7 channels promotes synaptic transmission and activates Akt1/GSK‐3β signalling.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| | - Wenwen Yu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China.,Institute of Innovative Drugs, Qingdao University, Qingdao, China
| | - Qin Gao
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| | - Chuanxia Ju
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| | - KeWei Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China.,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, China.,Institute of Innovative Drugs, Qingdao University, Qingdao, China
| |
Collapse
|
24
|
Wang J, Yang B, Ju L, Yang J, Allen A, Zhang J, Martynyuk AE. The Estradiol Synthesis Inhibitor Formestane Diminishes the Ability of Sevoflurane to Induce Neurodevelopmental Abnormalities in Male Rats. Front Syst Neurosci 2020; 14:546531. [PMID: 33013333 PMCID: PMC7498728 DOI: 10.3389/fnsys.2020.546531] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 08/14/2020] [Indexed: 01/14/2023] Open
Abstract
Background In rodents, the period of increased vulnerability to the developmental effects of general anesthetics coincides with the period of age-specific organizing (masculinizing) effects of the major female sex hormone 17β-estradiol (E2) in the male brain and excitatory GABA type A receptor (GABAAR) signaling. We studied whether E2 synthesis and excitatory GABAAR signaling are involved in the mediation of the developmental effects of sevoflurane in male rats. Methods Male Sprague-Dawley rats were pretreated with the inhibitors of E2 synthesis, formestane, or the Na+-K+-2Cl– (NKCC1) Cl– importer, bumetanide, prior to sevoflurane exposure for 6 h on postnatal (P) day 4, P5, or P6. We tested whether a subsequent exposure of these rats to sevoflurane on P∼10 would cause electroencephalography (EEG)-detectable seizures. We also evaluated their behavior during the elevated plus maze (EPM) test on P∼60, prepulse inhibition (PPI) of acoustic startle responses on P∼70, and corticosterone secretion to physical restraint on P∼80. Results The rats neonatally exposed to sevoflurane responded to repeated exposure to sevoflurane with increased EEG-detectable seizures (F(3,24) = 7.445, P = 0.001) and exhibited deficiencies during the EPM (F(3,55) = 4.397, P = 0.008) and PPI (F(3,110) = 5.222, P = 0.003) tests. They also responded to physical restraint with heightened secretion of corticosterone (F(3,16) = 11.906, P < 0.001). These parameters in the sevoflurane-exposed rats that were pretreated with formestane or bumetanide were not different from those in the control rats. Conclusion These results, along with previously published data, suggest that sevoflurane-enhanced E2 synthesis and excitatory GABAAR signaling at the time of sevoflurane anesthesia are involved in the mediation of the neurodevelopmental effects of the anesthetic in male rats.
Collapse
Affiliation(s)
- Jie Wang
- Department of Anesthesiology and Perioperative Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China.,Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL, United States
| | - Baofeng Yang
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL, United States.,Department of Anesthesiology and Perioperative Medicine, Affiliated, Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Lingsha Ju
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL, United States
| | - Jiaojiao Yang
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL, United States
| | - Andrea Allen
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL, United States
| | - Jiaqiang Zhang
- Department of Anesthesiology and Perioperative Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Anatoly E Martynyuk
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL, United States.,The McKnight Brain Institute, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
25
|
Martin-Kenny N, Bérubé NG. Effects of a postnatal Atrx conditional knockout in neurons on autism-like behaviours in male and female mice. J Neurodev Disord 2020; 12:17. [PMID: 32580781 PMCID: PMC7315487 DOI: 10.1186/s11689-020-09319-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 06/04/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Alpha-thalassemia/mental retardation, X-linked, or ATRX, is an autism susceptibility gene that encodes a chromatin remodeler. Mutations of ATRX result in the ATR-X intellectual disability syndrome and have been identified in autism spectrum disorder (ASD) patients. The mechanisms by which ATRX mutations lead to autism and autistic-like behaviours are not yet known. To address this question, we generated mice with postnatal Atrx inactivation in excitatory neurons of the forebrain and performed a battery of behavioural assays that assess autistic-like behaviours. METHODS Male and female mice with a postnatal conditional ablation of ATRX were generated using the Cre/lox system under the control of the αCaMKII gene promoter. These mice were tested in a battery of behavioural tests that assess autistic-like features. We utilized paradigms that measure social behaviour, repetitive, and stereotyped behaviours, as well as sensory gating. Statistics were calculated by two-way repeated measures ANOVA with Sidak's multiple comparison test or unpaired Student's t tests as indicated. RESULTS The behaviour tests revealed no significant differences between Atrx-cKO and control mice. We identified sexually dimorphic changes in odor habituation and discrimination; however, these changes did not correlate with social deficits. CONCLUSION The postnatal knockout of Atrx in forebrain excitatory neurons does not lead to autism-related behaviours in male or female mice.
Collapse
Affiliation(s)
- Nicole Martin-Kenny
- Department of Paediatrics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Division of Genetics and Development, Children's Health Research Institute, London, Ontario, Canada
| | - Nathalie G Bérubé
- Department of Paediatrics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
- Division of Genetics and Development, Children's Health Research Institute, London, Ontario, Canada.
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
| |
Collapse
|
26
|
Bahceci D, Anderson LL, Occelli Hanbury Brown CV, Zhou C, Arnold JC. Adolescent behavioral abnormalities in a Scn1a +/- mouse model of Dravet syndrome. Epilepsy Behav 2020; 103:106842. [PMID: 31870807 DOI: 10.1016/j.yebeh.2019.106842] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/03/2019] [Accepted: 12/03/2019] [Indexed: 12/30/2022]
Abstract
Dravet syndrome is an intractable pediatric epilepsy associated with SCN1A mutations. In addition to having a large seizure burden and a reduced lifespan, patients with Dravet syndrome also exhibit delays in reaching normal developmental milestones in attentional, emotional, and cognitive function. These developmental delays manifest in autistic-like social withdrawal and compulsive behavior. Additionally, cognitive impairments including deficits in sensorimotor processing and memory function are present. Several mouse models utilizing heterozygous deletion of Scn1a (Scn1a+/- mice) have been generated that recapitulate many aspects of Dravet syndrome. Studies in these mouse models of Dravet syndrome have characterized behavioral phenotypes in adult mice. In the present study, we characterized the behavioral phenotype of Scn1a+/- mice generated by targeted deletion of Scn1a exon 1 (Scn1atm1Kea) during adolescence. Identifying behavioral deficits in adolescent mice would more closely model the early onset of attentional, emotional, and cognitive delays observed in patients with Dravet syndrome. The behaviors of adolescent Scn1a+/- and wildtype (WT) mice were compared across several behavioral domains. We assessed motor function (open-field test), sociability and social recognition memory (three-chambered social preference and social interaction tests), memory function (novel object recognition, Barnes maze, fear conditioning paradigm), anxiety-related behavior (elevated plus maze and open-field thigmotaxis), startle reflex and sensorimotor gating (prepulse inhibition of startle (PPI) tests), and repetitive compulsive behavior (marble burying test). Adolescent Scn1a+/- mice exhibited normal locomotor activity, marble burying behavior, sociability, and sensorimotor gating. However, adolescent Scn1a+/- mice displayed increased anxiety-related thigmotactic behavior, atypical fear expression, blunted acoustic startle responses, and impaired social recognition and spatial memory. Our results show that Scn1a+/- mice display various behavioral impairments during adolescence, which provides a foundation for testing early intervention therapies targeting developmental delays modeled in Dravet syndrome mice.
Collapse
Affiliation(s)
- Dilara Bahceci
- Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia; School of Medical Science and Discipline of Pharmacology, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Lyndsey Leigh Anderson
- Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia; School of Medical Science and Discipline of Pharmacology, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Cassandra Veronica Occelli Hanbury Brown
- Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia; School of Medical Science and Discipline of Pharmacology, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Cilla Zhou
- Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia; School of Medical Science and Discipline of Pharmacology, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Jonathon Carl Arnold
- Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia; School of Medical Science and Discipline of Pharmacology, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
27
|
Fulcher N, Azzopardi E, De Oliveira C, Hudson R, Schormans AL, Zaman T, Allman BL, Laviolette SR, Schmid S. Deciphering midbrain mechanisms underlying prepulse inhibition of startle. Prog Neurobiol 2019; 185:101734. [PMID: 31863802 DOI: 10.1016/j.pneurobio.2019.101734] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/19/2019] [Accepted: 12/11/2019] [Indexed: 12/16/2022]
Abstract
Prepulse inhibition (PPI) is an operational measure of sensorimotor gating. Deficits of PPI are a hallmark of schizophrenia and associated with several other psychiatric illnesses such as e.g. autism spectrum disorder, yet the mechanisms underlying PPI are still not fully understood. There is growing evidence contradicting the long-standing hypothesis that PPI is mediated by a short feed-forward midbrain circuitry including inhibitory cholinergic projections from the pedunculopontine tegmental nucleus (PPTg) to the startle pathway. Here, we employed a chemogenetic approach to explore the involvement of the PPTg in general, and cholinergic neurons specifically, in PPI. Activation of inhibitory DREADDs (designer receptors exclusively activated by designer drugs) in the PPTg by systemic administration of clozapine-N-oxide (CNO) disrupted PPI, confirming the involvement of the PPTg in PPI. In contrast, chemogenetic inhibition of specifically cholinergic PPTg neurons had no effect on PPI, but inhibited morphine-induced conditioned place preference (CPP) in the same animals, showing that the DREADDs were effective in modulating behavior. These findings support a functional role of the PPTg and/or neighboring structures in PPI in accordance with previous lesion studies, but also provide strong evidence against the hypothesis that specifically cholinergic PPTg neurons are involved in mediating PPI, implicating rather non-cholinergic midbrain neurons.
Collapse
Affiliation(s)
- Niveen Fulcher
- University of Western Ontario, Schulich School of Medicine & Dentistry, Neuroscience Graduate Program, London, ON, N6A 5C1 Canada
| | - Erin Azzopardi
- University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada
| | - Cleusa De Oliveira
- University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada
| | - Roger Hudson
- University of Western Ontario, Schulich School of Medicine & Dentistry, Neuroscience Graduate Program, London, ON, N6A 5C1 Canada
| | - Ashley L Schormans
- University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada
| | - Tariq Zaman
- University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada
| | - Brian L Allman
- University of Western Ontario, Schulich School of Medicine & Dentistry, Neuroscience Graduate Program, London, ON, N6A 5C1 Canada; University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada
| | - Steven R Laviolette
- University of Western Ontario, Schulich School of Medicine & Dentistry, Neuroscience Graduate Program, London, ON, N6A 5C1 Canada; University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada
| | - Susanne Schmid
- University of Western Ontario, Schulich School of Medicine & Dentistry, Neuroscience Graduate Program, London, ON, N6A 5C1 Canada; University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada.
| |
Collapse
|
28
|
Moszczynski AJ, Harvey M, Fulcher N, de Oliveira C, McCunn P, Donison N, Bartha R, Schmid S, Strong MJ, Volkening K. Synergistic toxicity in an in vivo model of neurodegeneration through the co-expression of human TDP-43 M337V and tau T175D protein. Acta Neuropathol Commun 2019; 7:170. [PMID: 31703746 PMCID: PMC6839082 DOI: 10.1186/s40478-019-0816-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 09/22/2019] [Indexed: 02/08/2023] Open
Abstract
Although it has been suggested that the co-expression of multiple pathological proteins associated with neurodegeneration may act synergistically to induce more widespread neuropathology, experimental evidence of this is sparse. We have previously shown that the expression of Thr175Asp-tau (tauT175D) using somatic gene transfer with a stereotaxically-injected recombinant adeno-associated virus (rAAV9) vector induces tau pathology in rat hippocampus. In this study, we have examined whether the co-expression of human tauT175D with mutant human TDP-43 (TDP-43M337V) will act synergistically. Transgenic female Sprague-Dawley rats that inducibly express mutant human TDP-43M337V using the choline acetyltransferase (ChAT) tetracycline response element (TRE) driver with activity modulating tetracycline-controlled transactivator (tTA) were utilized in these studies. Adult rats were injected with GFP-tagged tau protein constructs in a rAAV9 vector through bilateral stereotaxic injection into the hippocampus. Injected tau constructs were: wild-type GFP-tagged 2N4R human tau (tauWT; n = 8), GFP-tagged tauT175D 2N4R human tau (tauT175D, pseudophosphorylated, toxic variant, n = 8), and GFP (control, n = 8). Six months post-injection, mutant TDP-43M337V expression was induced for 30 days. Behaviour testing identified motor deficits within 3 weeks after TDP-43 expression irrespective of tau expression, though social behaviour and sensorimotor gating remained unchanged. Increased tau pathology was observed in the hippocampus of both tauWT and tauT175D expressing rats and tauT175D pathology was increased in the presence of cholinergic neuronal expression of human TDP-43M337V. These data indicate that co-expression of pathological TDP-43 and tau protein exacerbate the pathology associated with either individual protein.
Collapse
|
29
|
Kulikova EA, Kulikov AV. Tryptophan hydroxylase 2 as a therapeutic target for psychiatric disorders: focus on animal models. Expert Opin Ther Targets 2019; 23:655-667. [PMID: 31216212 DOI: 10.1080/14728222.2019.1634691] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: Tryptophan hydroxylase 2 (TPH2) is the key, rate-limiting enzyme of serotonin (5-HT) synthesis in the brain. Some polymorphic variants of the human Tph2 gene are associated with psychiatric disorders. Area covered: This review focuses on the mechanisms underlying the association between the TPH2 activity and behavioral disturbances in models of psychiatric disorders. Specifically, it discusses: 1) genetic and posttranslational mechanisms defining the TPH2 activity, 2) behavioral effects of knockout and loss-of-function mutations in the mouse Tph2 gene, 3) pharmacological inhibition and the activation of the TPH2 activity and 4) alterations in the brain TPH2 activity in animal models of psychiatric disorders. We show the dual role of the TPH2 activity: both deficit and excess of the TPH2 activity cause significant behavioral disturbances in animal models of depression, anxiety, aggression, obsessive-compulsive disorders, schizophrenia, and catalepsy. Expert opinion: Pharmacological chaperones correcting the structure of the TPH2 molecule are promising tools for treatment of some hereditary psychiatric disorders caused by loss-of-function mutations in the human Tph2 gene; while some stress-induced affective disorders, associated with the elevated TPH2 activity, may be effectively treated by TPH2 inhibitors. This dual role of TPH2 should be taken into consideration during therapy of psychiatric disorders.
Collapse
Affiliation(s)
- Elizabeth A Kulikova
- a Federal Research Center Institute of Cytology and Genetics , Siberian Division of the Russian Academy of Science , Novosibirsk , Russia
| | - Alexander V Kulikov
- a Federal Research Center Institute of Cytology and Genetics , Siberian Division of the Russian Academy of Science , Novosibirsk , Russia
| |
Collapse
|
30
|
Hefter D, Topor CE, Gass P, Hirjak D. Two Sides of the Same Coin: A Case Report of First-Episode Catatonic Syndrome in a High-Functioning Autism Patient. Front Psychiatry 2019; 10:224. [PMID: 31031660 PMCID: PMC6473553 DOI: 10.3389/fpsyt.2019.00224] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 03/26/2019] [Indexed: 01/15/2023] Open
Abstract
Background: Catatonic phenomena such as stupor, mutism, stereotypy, echolalia, echopraxia, affective flattening, psychomotor deficits, and social withdrawal are characteristic symptoms of both schizophrenia and autism spectrum disorders (ASD), suggesting overlapping pathophysiological similarities such as altered glutamatergic and dopaminergic synaptic transmission and common genetic mutations. In daily clinical practice, ASD can be masked by manifest catatonic or psychotic symptoms and represent a diagnostic challenge, especially in patients with unknown or empty medical history. Unclear diagnosis is one of the main factors for delayed treatment. However, we are still missing diagnostic recommendations when dealing with ASD patients suffering from catatonic syndrome. Case presentation: A 31-year-old male patient without history of psychiatric disease presented with a severe catatonic syndrome and was admitted to our closed psychiatric ward. After the treatment with high-dose lorazepam and intramuscular olanzapine, catatonic symptoms largely remitted, but autistic traits persisted. Following a detailed anamnesis and a thorough neuropsychological testing, we diagnosed the patient with high-functioning autism and catatonic schizophrenia. The patient was discharged in a remitted state with long-acting injectable olanzapine. Conclusion: This case represents an example of diagnostic and therapeutic challenges of catatonic schizophrenia in high-functioning autism due to clinical and neurobiological overlaps of these conditions. We discuss clinical features together with pathophysiological concepts of both conditions. Furthermore, we tackle social and legal hurdles in Germany that naturally arise in these patients. Finally, we present diagnostic "red flags" that can be used to rationally select and conduct current recommended diagnostic assessments if there is a suspicion of ASD in patients with catatonic syndrome in order to provide them with the most appropriate treatment.
Collapse
Affiliation(s)
- Dimitri Hefter
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Cristina E. Topor
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Peter Gass
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Dusan Hirjak
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
31
|
Ebishima K, Takahashi H, Stickley A, Nakahachi T, Sumiyoshi T, Kamio Y. Relationship of the Acoustic Startle Response and Its Modulation to Adaptive and Maladaptive Behaviors in Typically Developing Children and Those With Autism Spectrum Disorders: A Pilot Study. Front Hum Neurosci 2019; 13:5. [PMID: 30723400 PMCID: PMC6349714 DOI: 10.3389/fnhum.2019.00005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/07/2019] [Indexed: 12/27/2022] Open
Abstract
Background: Autism spectrum disorder (ASD) is associated with persistent impairments in adaptive functioning across multiple domains of daily life. Thus, investigation of the biological background of both adaptive and maladaptive behaviors may shed light on developing effective interventions for improving social adaptation in ASD. In this study, we examined the relationship between adaptive/maladaptive behaviors and the acoustic startle response (ASR) and its modulation, which are promising neurophysiological markers for ASD translational research. Method: We investigated the ASR and its modulation in 11 children with ASD and 18 with typical development (TD), analyzing the relationship between startle measures and adaptive/maladaptive behaviors assessed with the Vineland Adaptive Behavior Scales (VABS) Second Edition. Results: Peak-ASR latency was negatively correlated with the VABS total score and socialization domain score of adaptive behaviors, while the ASR magnitude for relatively weak stimuli of 75–85 dB was positively correlated with VABS maladaptive behavior scores. Prepulse inhibition (PPI) at the prepulse intensity of 70–75 dB was also correlated with VABS maladaptive behavior. However, these relationships did not remain significant after adjustment for multiple comparisons. Conclusions: Our results indicate that the prolonged peak-ASR latency of ASD children might be associated with impairment in the developmental level of adaptive behavior, and that the greater ASR magnitude to relatively weak acoustic stimuli and smaller PPI of ASD children might increase the risk of maladaptive behavior. Future studies that have larger sample sizes will be important for further elucidating the neurophysiological factors that underpin adaptive as well as maladaptive behaviors in ASD.
Collapse
Affiliation(s)
- Ken Ebishima
- National Center of Neurology and Psychiatry, Department of Preventive Intervention for Psychiatric Disorders, National Institute of Mental Health, Tokyo, Japan
| | - Hidetoshi Takahashi
- National Center of Neurology and Psychiatry, Department of Preventive Intervention for Psychiatric Disorders, National Institute of Mental Health, Tokyo, Japan.,Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, Department of Advanced Neuroimaging, Tokyo, Japan
| | - Andrew Stickley
- National Center of Neurology and Psychiatry, Department of Preventive Intervention for Psychiatric Disorders, National Institute of Mental Health, Tokyo, Japan.,Stockholm Center for Health and Social Change (SCOHOST), Södertörn University, Huddinge, Sweden
| | - Takayuki Nakahachi
- National Center of Neurology and Psychiatry, Department of Preventive Intervention for Psychiatric Disorders, National Institute of Mental Health, Tokyo, Japan
| | - Tomiki Sumiyoshi
- National Center of Neurology and Psychiatry, Department of Preventive Intervention for Psychiatric Disorders, National Institute of Mental Health, Tokyo, Japan
| | - Yoko Kamio
- National Center of Neurology and Psychiatry, Department of Preventive Intervention for Psychiatric Disorders, National Institute of Mental Health, Tokyo, Japan
| |
Collapse
|
32
|
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: 29] [Impact Index Per Article: 4.1] [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
|
33
|
Imai K, Kotani T, Tsuda H, Nakano T, Ushida T, Iwase A, Nagai T, Toyokuni S, Suzumura A, Kikkawa F. Administration of molecular hydrogen during pregnancy improves behavioral abnormalities of offspring in a maternal immune activation model. Sci Rep 2018; 8:9221. [PMID: 29907804 PMCID: PMC6003913 DOI: 10.1038/s41598-018-27626-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 06/06/2018] [Indexed: 12/27/2022] Open
Abstract
The aim of the present study was to investigate long-term outcomes of the offspring in a lipopolysaccharide (LPS)-induced maternal immune activation (MIA) model and the effect of maternal molecular hydrogen (H2) administration. We have previously demonstrated in the MIA mouse model that maternal administration of H2 attenuates oxidative damage and neuroinflammation, including induced pro-inflammatory cytokines and microglial activation, in the fetal brain. Short-term memory, sociability and social novelty, and sensorimotor gating were evaluated using the Y-maze, three-chamber, and prepulse inhibition (PPI) tests, respectively, at postnatal 3 or 4 weeks. The number of neurons and oligodendrocytes was also analyzed at postnatal 5 weeks by immunohistochemical analysis. Offspring of the LPS-exposed dams showed deficits in short-term memory and social interaction, following neuronal and oligodendrocytic loss in the amygdala and cortex. Maternal H2 administration markedly attenuated these LPS-induced abnormalities. Moreover, we evaluated the effect of H2 on LPS-induced astrocytic activation, both in vivo and in vitro. The number of activated astrocytes with hypertrophic morphology was increased in LPS-exposed offspring, but decreased in the offspring of H2-administered dams. In primary cultured astrocytes, LPS-induced pro-inflammatory cytokines were attenuated by H2 administration. Overall, these findings indicate that maternal H2 administration exerts neuroprotective effects and ameliorates MIA-induced neurodevelopmental deficits of offspring later in life.
Collapse
Affiliation(s)
- Kenji Imai
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Tomomi Kotani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Hiroyuki Tsuda
- Department of Obstetrics and Gynecology, Japanese Red Cross Nagoya Daiichi Hospital, 3-35, Michishita-Cho, Nakamura-Ku, Nagoya, 453-8511, Japan
| | - Tomoko Nakano
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Takafumi Ushida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Akira Iwase
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Taku Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Akio Suzumura
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601, Japan
| | - Fumitaka Kikkawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
34
|
Takahashi H, Nakamura T, Kim J, Kikuchi H, Nakahachi T, Ishitobi M, Ebishima K, Yoshiuchi K, Ando T, Stickley A, Yamamoto Y, Kamio Y. Acoustic Hyper-Reactivity and Negatively Skewed Locomotor Activity in Children With Autism Spectrum Disorders: An Exploratory Study. Front Psychiatry 2018; 9:355. [PMID: 30127755 PMCID: PMC6088201 DOI: 10.3389/fpsyt.2018.00355] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 07/16/2018] [Indexed: 12/27/2022] Open
Abstract
Investigation of objective and quantitative behavioral phenotypes along with neurobiological endophenotypes might lead to increased knowledge of the mechanisms that underlie autism spectrum disorders (ASD). Here, we investigated the association between locomotor dynamics and characteristics of the acoustic startle response (ASR) and its modulation in ASD (n = 14) and typically developing (TD, n = 13) children. The ASR was recorded in response to acoustic stimuli in increments of 10 dB (65-105 dB SPL). We calculated the average ASR magnitude for each stimulus intensity and peak-ASR latency. Locomotor activity was continuously measured with a watch-type actigraph. We examined statistics of locomotor activity, such as mean activity levels and the skewness of activity. Children with ASD had a significantly greater ASR magnitude in response to a weak acoustic stimulus, which reflects acoustic hyper-reactivity. The skewness of all-day activity was significantly more negative in children with ASD than those with TD. Skewness of daytime activity was also more negative, although only of borderline statistical significance. For all children, the higher mean and more negatively skewed daytime activity, reflecting hyperactivity that was associated with sporadic large daytime "troughs," was significantly correlated with acoustic hyper-reactivity. The more negatively skewed locomotor activity occurring in the daytime was also associated with impaired sensorimotor gating, examined as prepulse inhibition at a prepulse intensity of 70 dB. This comprehensive investigation of locomotor dynamics and the ASR extends our understanding of the neurophysiology that underlies ASD.
Collapse
Affiliation(s)
- Hidetoshi Takahashi
- Department of Child and Adolescent Mental Health, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan.,Department of Advanced Neuroimaging, Integrative Brain Imaging Centerm, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Toru Nakamura
- Graduate School of Education, The University of Tokyo, 2Tokyo, Japan
| | - Jinhyuk Kim
- Graduate School of Education, The University of Tokyo, 2Tokyo, Japan
| | - Hiroe Kikuchi
- Department of Psychosomatic Research, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Takayuki Nakahachi
- Department of Child and Adolescent Mental Health, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Makoto Ishitobi
- Department of Child and Adolescent Mental Health, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Ken Ebishima
- Department of Child and Adolescent Mental Health, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Kazuhiro Yoshiuchi
- Department of Stress Sciences and Psychosomatic Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Tetsuya Ando
- Department of Psychosomatic Research, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Andrew Stickley
- Department of Child and Adolescent Mental Health, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan.,Stockholm Center for Health and Social Change, Södertörn University, Huddinge, Sweden
| | | | - Yoko Kamio
- Department of Child and Adolescent Mental Health, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|