1
|
Vanessa Becerra-Hernández L, Casanova MF, Buriticá E. Cortical calretinin-positive neurons: Functional and ontogenetic characteristics and their relationship to brain pathologies. Brain Res 2024; 1846:149285. [PMID: 39490954 DOI: 10.1016/j.brainres.2024.149285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 06/02/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024]
Abstract
Cortical GABAergic interneurons can be classified according to electrophysiological, biochemical, and/or morphological criteria. In humans, the use of calcium-binding proteins allows us to differentiate three subpopulations of GABAergic interneurons with minimal overlap. Cortical calretinin-positive neurons mainly include bipolar and double-bouquet morphologies, with a largely non-rapid and adaptive firing pattern, originating from the ganglionic eminence and the ventricular and subventricular regions of the developing brain. These cells are distributed from layer I to VI of the neocortex, with predominance in layers II and III. Given their morphology, distribution of processes, and elucidated synaptic contacts, these neurons are considered important in the control of intraminicolumnar processing through vertical inhibition. They have been extensively studied in the context of pathologies characterized by excitation/inhibition imbalance, such as Alzheimer's disease, epilepsy, traumatic brain injury, and autism. In light of the current evidence, this review considers these aspects in depth and discusses the pathophysiological role and selective vulnerability (pathoclisis) vs. the resistance that these interneurons can present against different types of injury.
Collapse
Affiliation(s)
- Lina Vanessa Becerra-Hernández
- Centro de Estudios Cerebrales, Facultad de Salud, Universidad del Valle, Cali, Colombia; Departamento de Ciencias Básicas de la Salud, Pontificia Universidad Javeriana, Cali, Colombia.
| | - Manuel F Casanova
- Department of Biomedical Sciences, School of Medicine Greenville, University of South Carolina, Greenville, SC, United States
| | - Efraín Buriticá
- Centro de Estudios Cerebrales, Facultad de Salud, Universidad del Valle, Cali, Colombia
| |
Collapse
|
2
|
Souza LRQ, Pedrosa CGDS, Puig-Pijuan T, da Silva Dos Santos C, Vitória G, Delou JMA, Setti-Perdigão P, Higa LM, Tanuri A, Rehen SK, Guimarães MZP. Saxitoxin potentiates human neuronal cell death induced by Zika virus while sparing neural progenitors and astrocytes. Sci Rep 2024; 14:22809. [PMID: 39354036 PMCID: PMC11445263 DOI: 10.1038/s41598-024-73873-z] [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: 06/21/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Abstract
The Zika virus (ZIKV) epidemic declared in Brazil between 2015 and 2016 was associated with an increased prevalence of severe congenital malformations, including microcephaly. The distribution of microcephaly cases was not uniform across the country, with a disproportionately higher incidence in the Northeast region (NE). Our previous work demonstrated that saxitoxin (STX), a toxin present in the drinking water reservoirs of the NE, exacerbated the damaging effects of ZIKV on the developing brain. We hypothesized that the impact of STX might vary among different neural cell types. While ZIKV infection caused severe damages on astrocytes and neural stem cells (NSCs), the addition of STX did not exacerbate these effects. We observed that neurons subjected to STX exposure were more prone to apoptosis and displayed higher ZIKV infection rate. These findings suggest that STX exacerbates the harmful effects of ZIKV on neurons, thereby providing a plausible explanation for the heightened severity of ZIKV-induced congenital malformations observed in Brazil's NE. This study highlights the importance of understanding the interactive effects of environmental toxins and infectious pathogens on neural development, with potential implications for public health policies.
Collapse
Affiliation(s)
- Leticia R Q Souza
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
| | - Carolina G da S Pedrosa
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
| | - Teresa Puig-Pijuan
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
| | | | - Gabriela Vitória
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
| | - João M A Delou
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
| | - Pedro Setti-Perdigão
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
| | - Luiza M Higa
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Amilcar Tanuri
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Stevens Kastrup Rehen
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Marília Zaluar P Guimarães
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil.
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.
| |
Collapse
|
3
|
Dorsey SG, Mocci E, Lane MV, Krueger BK. Rapid effects of valproic acid on the fetal brain transcriptome: Implications for brain development and autism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.01.538959. [PMID: 37205520 PMCID: PMC10187231 DOI: 10.1101/2023.05.01.538959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
There is an increased incidence of autism among the children of women who take the anti-epileptic, mood stabilizing drug, valproic acid (VPA) during pregnancy; moreover, exposure to VPA in utero causes autistic-like symptoms in rodents and non-human primates. Analysis of RNAseq data obtained from fetal mouse brains 3 hr after VPA administration revealed that VPA significantly [p(FDR) ≤ 0.025] increased or decreased the expression of approximately 7,300 genes. No significant sex differences in VPA-induced gene expression were observed. Expression of genes associated with neurodevelopmental disorders such as autism as well as neurogenesis, axon growth and synaptogenesis, GABAergic, glutaminergic and dopaminergic synaptic transmission, perineuronal nets, and circadian rhythms was dysregulated by VPA. Moreover, expression of 400 autism risk genes was significantly altered by VPA as was expression of 247 genes that have been reported to play fundamental roles in the development of the nervous system, but are not linked to autism by GWAS. The goal of this study was to identify mouse genes that are: (a) significantly up- or down-regulated by VPA in the fetal brain and (b) known to be associated with autism and/or to play a role in embryonic neurodevelopmental processes, perturbation of which has the potential to alter brain connectivity in the postnatal and adult brain. The set of genes meeting these criteria provides potential targets for future hypothesis-driven approaches to elucidating the proximal underlying causes of defective brain connectivity in neurodevelopmental disorders such as autism.
Collapse
|
4
|
Transition from Animal-Based to Human Induced Pluripotent Stem Cells (iPSCs)-Based Models of Neurodevelopmental Disorders: Opportunities and Challenges. Cells 2023; 12:cells12040538. [PMID: 36831205 PMCID: PMC9954744 DOI: 10.3390/cells12040538] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/25/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Neurodevelopmental disorders (NDDs) arise from the disruption of highly coordinated mechanisms underlying brain development, which results in impaired sensory, motor and/or cognitive functions. Although rodent models have offered very relevant insights to the field, the translation of findings to clinics, particularly regarding therapeutic approaches for these diseases, remains challenging. Part of the explanation for this failure may be the genetic differences-some targets not being conserved between species-and, most importantly, the differences in regulation of gene expression. This prompts the use of human-derived models to study NDDS. The generation of human induced pluripotent stem cells (hIPSCs) added a new suitable alternative to overcome species limitations, allowing for the study of human neuronal development while maintaining the genetic background of the donor patient. Several hIPSC models of NDDs already proved their worth by mimicking several pathological phenotypes found in humans. In this review, we highlight the utility of hIPSCs to pave new paths for NDD research and development of new therapeutic tools, summarize the challenges and advances of hIPSC-culture and neuronal differentiation protocols and discuss the best way to take advantage of these models, illustrating this with examples of success for some NDDs.
Collapse
|
5
|
Lee K, Mills Z, Cheung P, Cheyne JE, Montgomery JM. The Role of Zinc and NMDA Receptors in Autism Spectrum Disorders. Pharmaceuticals (Basel) 2022; 16:ph16010001. [PMID: 36678498 PMCID: PMC9866730 DOI: 10.3390/ph16010001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
NMDA-type glutamate receptors are critical for synaptic plasticity in the central nervous system. Their unique properties and age-dependent arrangement of subunit types underpin their role as a coincidence detector of pre- and postsynaptic activity during brain development and maturation. NMDAR function is highly modulated by zinc, which is co-released with glutamate and concentrates in postsynaptic spines. Both NMDARs and zinc have been strongly linked to autism spectrum disorders (ASDs), suggesting that NMDARs are an important player in the beneficial effects observed with zinc in both animal models and children with ASDs. Significant evidence is emerging that these beneficial effects occur via zinc-dependent regulation of SHANK proteins, which form the backbone of the postsynaptic density. For example, dietary zinc supplementation enhances SHANK2 or SHANK3 synaptic recruitment and rescues NMDAR deficits and hypofunction in Shank3ex13-16-/- and Tbr1+/- ASD mice. Across multiple studies, synaptic changes occur in parallel with a reversal of ASD-associated behaviours, highlighting the zinc-dependent regulation of NMDARs and glutamatergic synapses as therapeutic targets for severe forms of ASDs, either pre- or postnatally. The data from rodent models set a strong foundation for future translational studies in human cells and people affected by ASDs.
Collapse
|
6
|
Reddy DS, Abeygunaratne HN. Experimental and Clinical Biomarkers for Progressive Evaluation of Neuropathology and Therapeutic Interventions for Acute and Chronic Neurological Disorders. Int J Mol Sci 2022; 23:11734. [PMID: 36233034 PMCID: PMC9570151 DOI: 10.3390/ijms231911734] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/27/2022] Open
Abstract
This article describes commonly used experimental and clinical biomarkers of neuronal injury and neurodegeneration for the evaluation of neuropathology and monitoring of therapeutic interventions. Biomarkers are vital for diagnostics of brain disease and therapeutic monitoring. A biomarker can be objectively measured and evaluated as a proxy indicator for the pathophysiological process or response to therapeutic interventions. There are complex hurdles in understanding the molecular pathophysiology of neurological disorders and the ability to diagnose them at initial stages. Novel biomarkers for neurological diseases may surpass these issues, especially for early identification of disease risk. Validated biomarkers can measure the severity and progression of both acute neuronal injury and chronic neurological diseases such as epilepsy, migraine, Alzheimer's disease, Parkinson's disease, Huntington's disease, traumatic brain injury, amyotrophic lateral sclerosis, multiple sclerosis, and other brain diseases. Biomarkers are deployed to study progression and response to treatment, including noninvasive imaging tools for both acute and chronic brain conditions. Neuronal biomarkers are classified into four core subtypes: blood-based, immunohistochemical-based, neuroimaging-based, and electrophysiological biomarkers. Neuronal conditions have progressive stages, such as acute injury, inflammation, neurodegeneration, and neurogenesis, which can serve as indices of pathological status. Biomarkers are critical for the targeted identification of specific molecules, cells, tissues, or proteins that dramatically alter throughout the progression of brain conditions. There has been tremendous progress with biomarkers in acute conditions and chronic diseases affecting the central nervous system.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
- Institute of Pharmacology and Neurotherapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
- Intercollegiate School of Engineering Medicine, Texas A&M University, Houston, TX 77030, USA
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Hasara Nethma Abeygunaratne
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
- Institute of Pharmacology and Neurotherapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| |
Collapse
|
7
|
Crespo I, Pignatelli J, Kinare V, Méndez-Gómez HR, Esgleas M, Román MJ, Canals JM, Tole S, Vicario C. Tbr1 Misexpression Alters Neuronal Development in the Cerebral Cortex. Mol Neurobiol 2022; 59:5750-5765. [PMID: 35781633 PMCID: PMC9395452 DOI: 10.1007/s12035-022-02936-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 06/10/2022] [Indexed: 11/26/2022]
Abstract
Changes in the transcription factor (TF) expression are critical for brain development, and they may also underlie neurodevelopmental disorders. Indeed, T-box brain1 (Tbr1) is a TF crucial for the formation of neocortical layer VI, and mutations and microdeletions in that gene are associated with malformations in the human cerebral cortex, alterations that accompany autism spectrum disorder (ASD). Interestingly, Tbr1 upregulation has also been related to the occurrence of ASD-like symptoms, although limited studies have addressed the effect of increased Tbr1 levels during neocortical development. Here, we analysed the impact of Tbr1 misexpression in mouse neural progenitor cells (NPCs) at embryonic day 14.5 (E14.5), when they mainly generate neuronal layers II-IV. By E18.5, cells accumulated in the intermediate zone and in the deep cortical layers, whereas they became less abundant in the upper cortical layers. In accordance with this, the proportion of Sox5+ cells in layers V-VI increased, while that of Cux1+ cells in layers II-IV decreased. On postnatal day 7, fewer defects in migration were evident, although a higher proportion of Sox5+ cells were seen in the upper and deep layers. The abnormal neuronal migration could be partially due to the altered multipolar-bipolar neuron morphologies induced by Tbr1 misexpression, which also reduced dendrite growth and branching, and disrupted the corpus callosum. Our results indicate that Tbr1 misexpression in cortical NPCs delays or disrupts neuronal migration, neuronal specification, dendrite development and the formation of the callosal tract. Hence, genetic changes that provoke ectopic Tbr1 upregulation during development could provoke cortical brain malformations.
Collapse
Affiliation(s)
- Inmaculada Crespo
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- CES Cardenal Cisneros, Madrid, Spain
| | - Jaime Pignatelli
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Veena Kinare
- Department of Life Sciences, Sophia College for Women, Mumbai, 400026, India
| | - Héctor R Méndez-Gómez
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Miriam Esgleas
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, Creatio, Production and Validation Center of Advanced Therapies, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - María José Román
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Josep M Canals
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, Creatio, Production and Validation Center of Advanced Therapies, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Shubha Tole
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, 400005, India
| | - Carlos Vicario
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain.
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| |
Collapse
|
8
|
Zhang W, Hu L, Huang X, Xie D, Wu J, Fu X, Liang D, Huang S. Whole-exome sequencing identified five novel de novo variants in patients with unexplained intellectual disability. J Clin Lab Anal 2022; 36:e24587. [PMID: 35837997 PMCID: PMC9459325 DOI: 10.1002/jcla.24587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/20/2022] [Accepted: 06/25/2022] [Indexed: 11/30/2022] Open
Abstract
Background Intellectual disability (ID) represents a neurodevelopmental disorder, which is characterized by marked defects in the intellectual function and adaptive behavior, with an onset during the developmental period. ID is mainly caused by genetic factors, and it is extremely genetically heterogeneous. This study aims to identify the genetic cause of ID using trio‐WES analysis. Methods We recruited four pediatric patients with unexplained ID from non‐consanguineous families, who presented at the Department of Pediatrics, Guizhou Provincial People's Hospital. Whole‐exome sequencing (WES) and Sanger sequencing validation were performed in the patients and their unaffected parents. Furthermore, conservative analysis and protein structural and functional prediction were performed on the identified pathogenic variants. Results We identified five novel de novo mutations from four known ID‐causing genes in the four included patients, namely COL4A1 (c.2786T>A, p.V929D and c.2797G>A, p.G933S), TBR1 (c.1639_1640insCCCGCAGTCC, p.Y553Sfs*124), CHD7 (c.7013A>T, p.Q2338L), and TUBA1A (c.1350del, p.E450Dfs*34). These mutations were all predicted to be deleterious and were located at highly conserved domains that might affect the structure and function of these proteins. Conclusion Our findings contribute to expanding the mutational spectrum of ID‐related genes and help to deepen the understanding of the genetic causes and heterogeneity of ID.
Collapse
Affiliation(s)
- Wenqiu Zhang
- School of Medicine, Guizhou University, Guiyang, China.,Prenatal Diagnosis Center, Guizhou Provincial People's hospital, Guiyang, China
| | - Li Hu
- Prenatal Diagnosis Center, Guizhou Provincial People's hospital, Guiyang, China
| | - Xinyi Huang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dan Xie
- School of Medicine, Guizhou University, Guiyang, China.,Prenatal Diagnosis Center, Guizhou Provincial People's hospital, Guiyang, China
| | - Jiangfen Wu
- School of Medicine, Guizhou University, Guiyang, China.,Prenatal Diagnosis Center, Guizhou Provincial People's hospital, Guiyang, China
| | - Xiaoling Fu
- Department of Pediatrics, Guizhou Provincial People's hospital, Guiyang, China
| | - Daiyi Liang
- Department of Neurology, Guizhou Provincial People's hospital, Guiyang, China
| | - Shengwen Huang
- School of Medicine, Guizhou University, Guiyang, China.,Prenatal Diagnosis Center, Guizhou Provincial People's hospital, Guiyang, China.,NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
9
|
Slota JA, Medina SJ, Frost KL, Booth SA. Neurons and Astrocytes Elicit Brain Region Specific Transcriptional Responses to Prion Disease in the Murine CA1 and Thalamus. Front Neurosci 2022; 16:918811. [PMID: 35651626 PMCID: PMC9149297 DOI: 10.3389/fnins.2022.918811] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 04/29/2022] [Indexed: 01/14/2023] Open
Abstract
Progressive dysfunction and loss of neurons ultimately culminates in the symptoms and eventual fatality of prion disease, yet the pathways and mechanisms that lead to neuronal degeneration remain elusive. Here, we used RNAseq to profile transcriptional changes in microdissected CA1 and thalamus brain tissues from prion infected mice. Numerous transcripts were altered during clinical disease, whereas very few transcripts were reliably altered at pre-clinical time points. Prion altered transcripts were assigned to broadly defined brain cell types and we noted a strong transcriptional signature that was affiliated with reactive microglia and astrocytes. While very few neuronal transcripts were common between the CA1 and thalamus, we described transcriptional changes in both regions that were related to synaptic dysfunction. Using transcriptional profiling to compare how different neuronal populations respond during prion disease may help decipher mechanisms that lead to neuronal demise and should be investigated with greater detail.
Collapse
Affiliation(s)
- Jessy A. Slota
- One Health Division, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Sarah J. Medina
- One Health Division, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Kathy L. Frost
- One Health Division, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Stephanie A. Booth
- One Health Division, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
10
|
Lee K, Jung Y, Vyas Y, Skelton I, Abraham WC, Hsueh YP, Montgomery JM. Dietary zinc supplementation rescues fear-based learning and synaptic function in the Tbr1 +/- mouse model of autism spectrum disorders. Mol Autism 2022; 13:13. [PMID: 35303947 PMCID: PMC8932001 DOI: 10.1186/s13229-022-00494-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/07/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterised by a dyad of behavioural symptoms-social and communication deficits and repetitive behaviours. Multiple aetiological genetic and environmental factors have been identified as causing or increasing the likelihood of ASD, including serum zinc deficiency. Our previous studies revealed that dietary zinc supplementation can normalise impaired social behaviours, excessive grooming, and heightened anxiety in a Shank3 mouse model of ASD, as well as the amelioration of synapse dysfunction. Here, we have examined the efficacy and breadth of dietary zinc supplementation as an effective therapeutic strategy utilising a non-Shank-related mouse model of ASD-mice with Tbr1 haploinsufficiency. METHODS We performed behavioural assays, amygdalar slice whole-cell patch-clamp electrophysiology, and immunohistochemistry to characterise the synaptic mechanisms underlying the ASD-associated behavioural deficits observed in Tbr1+/- mice and the therapeutic potential of dietary zinc supplementation. Two-way analysis of variance (ANOVA) with Šídák's post hoc test and one-way ANOVA with Tukey's post hoc multiple comparisons were performed for statistical analysis. RESULTS Our data show that dietary zinc supplementation prevents impairments in auditory fear memory and social interaction, but not social novelty, in the Tbr1+/- mice. Tbr1 haploinsufficiency did not induce excessive grooming nor elevate anxiety in mice. At the synaptic level, dietary zinc supplementation reversed α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) and N-methyl-D-aspartate receptor (NMDAR) hypofunction and normalised presynaptic function at thalamic-lateral amygdala (LA) synapses that are crucial for auditory fear memory. In addition, the zinc supplemented diet significantly restored the synaptic puncta density of the GluN1 subunit essential for functional NMDARs as well as SHANK3 expression in both the basal and lateral amygdala (BLA) of Tbr1+/- mice. LIMITATIONS The therapeutic effect of dietary zinc supplementation observed in rodent models may not reproduce the same effects in human patients. The effect of dietary zinc supplementation on synaptic function in other brain structures affected by Tbr1 haploinsufficiency including olfactory bulb and anterior commissure will also need to be examined. CONCLUSIONS Our data further the understanding of the molecular mechanisms underlying the effect of dietary zinc supplementation and verify the efficacy and breadth of its application as a potential treatment strategy for ASD.
Collapse
Affiliation(s)
- Kevin Lee
- Department of Physiology and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Yewon Jung
- Department of Physiology and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Yukti Vyas
- Department of Physiology and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
- INSERM, Neurocentre Magendie, U1215, Bordeaux, France
| | - Imogen Skelton
- Department of Physiology and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Wickliffe C Abraham
- Department of Psychology and Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica, 128, Section 2, Academia Rd., Taipei, 11529, Taiwan
| | - Johanna M Montgomery
- Department of Physiology and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand.
| |
Collapse
|
11
|
Tocco C, Bertacchi M, Studer M. Structural and Functional Aspects of the Neurodevelopmental Gene NR2F1: From Animal Models to Human Pathology. Front Mol Neurosci 2022; 14:767965. [PMID: 34975398 PMCID: PMC8715095 DOI: 10.3389/fnmol.2021.767965] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/25/2021] [Indexed: 01/28/2023] Open
Abstract
The assembly and maturation of the mammalian brain result from an intricate cascade of highly coordinated developmental events, such as cell proliferation, migration, and differentiation. Any impairment of this delicate multi-factorial process can lead to complex neurodevelopmental diseases, sharing common pathogenic mechanisms and molecular pathways resulting in multiple clinical signs. A recently described monogenic neurodevelopmental syndrome named Bosch-Boonstra-Schaaf Optic Atrophy Syndrome (BBSOAS) is caused by NR2F1 haploinsufficiency. The NR2F1 gene, coding for a transcriptional regulator belonging to the steroid/thyroid hormone receptor superfamily, is known to play key roles in several brain developmental processes, from proliferation and differentiation of neural progenitors to migration and identity acquisition of neocortical neurons. In a clinical context, the disruption of these cellular processes could underlie the pathogenesis of several symptoms affecting BBSOAS patients, such as intellectual disability, visual impairment, epilepsy, and autistic traits. In this review, we will introduce NR2F1 protein structure, molecular functioning, and expression profile in the developing mouse brain. Then, we will focus on Nr2f1 several functions during cortical development, from neocortical area and cell-type specification to maturation of network activity, hippocampal development governing learning behaviors, assembly of the visual system, and finally establishment of cortico-spinal descending tracts regulating motor execution. Whenever possible, we will link experimental findings in animal or cellular models to corresponding features of the human pathology. Finally, we will highlight some of the unresolved questions on the diverse functions played by Nr2f1 during brain development, in order to propose future research directions. All in all, we believe that understanding BBSOAS mechanisms will contribute to further unveiling pathophysiological mechanisms shared by several neurodevelopmental disorders and eventually lead to effective treatments.
Collapse
Affiliation(s)
- Chiara Tocco
- Université Côte d'Azur, CNRS, Inserm, iBV, Nice, France
| | | | | |
Collapse
|
12
|
Liu Q, Zhang Y, Ouyang Z. Structural inference of time‐varying mixed graphical models. Stat (Int Stat Inst) 2021. [DOI: 10.1002/sta4.414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Qingyang Liu
- Department of Statistics University of Connecticut Storrs CT 06269 USA
| | - Yuping Zhang
- Department of Statistics University of Connecticut Storrs CT 06269 USA
| | - Zhengqing Ouyang
- Department of Biostatistics and Epidemiology School of Public Health and Health Sciences, University of Massachusetts Amherst Amherst MA 01003 USA
| |
Collapse
|
13
|
Yamagishi S, Bando Y, Sato K. Involvement of Netrins and Their Receptors in Neuronal Migration in the Cerebral Cortex. Front Cell Dev Biol 2021; 8:590009. [PMID: 33520982 PMCID: PMC7843923 DOI: 10.3389/fcell.2020.590009] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022] Open
Abstract
In mammals, excitatory cortical neurons develop from the proliferative epithelium and progenitor cells in the ventricular zone and subventricular zone, and migrate radially to the cortical plate, whereas inhibitory GABAergic interneurons are born in the ganglionic eminence and migrate tangentially. The migration of newly born cortical neurons is tightly regulated by both extracellular and intracellular signaling to ensure proper positioning and projections. Non-cell-autonomous extracellular molecules, such as growth factors, axon guidance molecules, extracellular matrix, and other ligands, play a role in cortical migration, either by acting as attractants or repellents. In this article, we review the guidance molecules that act as cell-cell recognition molecules for the regulation of neuronal migration, with a focus on netrin family proteins, their receptors, and related molecules, including neogenin, repulsive guidance molecules (RGMs), Down syndrome cell adhesion molecule (DSCAM), fibronectin leucine-rich repeat transmembrane proteins (FLRTs), and draxin. Netrin proteins induce attractive and repulsive signals depending on their receptors. For example, binding of netrin-1 to deleted in colorectal cancer (DCC), possibly together with Unc5, repels migrating GABAergic neurons from the ventricular zone of the ganglionic eminence, whereas binding to α3β1 integrin promotes cortical interneuron migration. Human genetic disorders associated with these and related guidance molecules, such as congenital mirror movements, schizophrenia, and bipolar disorder, are also discussed.
Collapse
Affiliation(s)
- Satoru Yamagishi
- Department of Organ and Tissue Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuki Bando
- Department of Organ and Tissue Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kohji Sato
- Department of Organ and Tissue Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
14
|
Prem S, Millonig JH, DiCicco-Bloom E. Dysregulation of Neurite Outgrowth and Cell Migration in Autism and Other Neurodevelopmental Disorders. ADVANCES IN NEUROBIOLOGY 2020; 25:109-153. [PMID: 32578146 DOI: 10.1007/978-3-030-45493-7_5] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Despite decades of study, elucidation of the underlying etiology of complex developmental disorders such as autism spectrum disorder (ASD), schizophrenia (SCZ), intellectual disability (ID), and bipolar disorder (BPD) has been hampered by the inability to study human neurons, the heterogeneity of these disorders, and the relevance of animal model systems. Moreover, a majority of these developmental disorders have multifactorial or idiopathic (unknown) causes making them difficult to model using traditional methods of genetic alteration. Examination of the brains of individuals with ASD and other developmental disorders in both post-mortem and MRI studies shows defects that are suggestive of dysregulation of embryonic and early postnatal development. For ASD, more recent genetic studies have also suggested that risk genes largely converge upon the developing human cerebral cortex between weeks 8 and 24 in utero. Yet, an overwhelming majority of studies in autism rodent models have focused on postnatal development or adult synaptic transmission defects in autism related circuits. Thus, studies looking at early developmental processes such as proliferation, cell migration, and early differentiation, which are essential to build the brain, are largely lacking. Yet, interestingly, a few studies that did assess early neurodevelopment found that alterations in brain structure and function associated with neurodevelopmental disorders (NDDs) begin as early as the initial formation and patterning of the neural tube. By the early to mid-2000s, the derivation of human embryonic stem cells (hESCs) and later induced pluripotent stem cells (iPSCs) allowed us to study living human neural cells in culture for the first time. Specifically, iPSCs gave us the unprecedented ability to study cells derived from individuals with idiopathic disorders. Studies indicate that iPSC-derived neural cells, whether precursors or "matured" neurons, largely resemble cortical cells of embryonic humans from weeks 8 to 24. Thus, these cells are an excellent model to study early human neurodevelopment, particularly in the context of genetically complex diseases. Indeed, since 2011, numerous studies have assessed developmental phenotypes in neurons derived from individuals with both genetic and idiopathic forms of ASD and other NDDs. However, while iPSC-derived neurons are fetal in nature, they are post-mitotic and thus cannot be used to study developmental processes that occur before terminal differentiation. Moreover, it is important to note that during the 8-24-week window of human neurodevelopment, neural precursor cells are actively undergoing proliferation, migration, and early differentiation to form the basic cytoarchitecture of the brain. Thus, by studying NPCs specifically, we could gain insight into how early neurodevelopmental processes contribute to the pathogenesis of NDDs. Indeed, a few studies have explored NPC phenotypes in NDDs and have uncovered dysregulations in cell proliferation. Yet, few studies have explored migration and early differentiation phenotypes of NPCs in NDDs. In this chapter, we will discuss cell migration and neurite outgrowth and the role of these processes in neurodevelopment and NDDs. We will begin by reviewing the processes that are important in early neurodevelopment and early cortical development. We will then delve into the roles of neurite outgrowth and cell migration in the formation of the brain and how errors in these processes affect brain development. We also provide review of a few key molecules that are involved in the regulation of neurite outgrowth and migration while discussing how dysregulations in these molecules can lead to abnormalities in brain structure and function thereby highlighting their contribution to pathogenesis of NDDs. Then we will discuss whether neurite outgrowth, migration, and the molecules that regulate these processes are associated with ASD. Lastly, we will review the utility of iPSCs in modeling NDDs and discuss future goals for the study of NDDs using this technology.
Collapse
Affiliation(s)
- Smrithi Prem
- Graduate Program in Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - James H Millonig
- Department of Neuroscience and Cell Biology, Center for Advanced Biotechnology and Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Emanuel DiCicco-Bloom
- Department of Neuroscience and Cell Biology/Pediatrics, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
15
|
Sapey-Triomphe LA, Reversat J, Lesca G, Chatron N, Bussa M, Mazoyer S, Schmitz C, Sonié S, Edery P. A de novo frameshift pathogenic variant in TBR1 identified in autism without intellectual disability. Hum Genomics 2020; 14:32. [PMID: 32948248 PMCID: PMC7501624 DOI: 10.1186/s40246-020-00281-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 08/31/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND In order to be able to provide accurate genetic counseling to patients with Autism Spectrum Disorder (ASD), it is crucial to identify correlations between heterogeneous phenotypes and genetic alterations. Among the hundreds of de novo pathogenic variants reported in ASD, single-nucleotide variations and small insertions/deletions were reported in TBR1. This gene encodes a transcription factor that plays a key role in brain development. Pathogenic variants in TBR1 are often associated with severe forms of ASD, including intellectual disability and language impairment. METHODS Adults diagnosed with ASD but without intellectual disability (diagnosis of Asperger syndrome, according to the DSM-IV) took part in a genetic consultation encompassing metabolic assessments, a molecular karyotype and the screening of a panel of 268 genes involved in intellectual disability, ASD and epilepsy. In addition, the patient reported here went through a neuropsychological assessment, structural magnetic resonance imaging and magnetic resonance spectroscopy measurements. RESULTS Here, we report the case of a young adult male who presents with a typical form of ASD. Importantly, this patient presents with no intellectual disability or language impairment, despite a de novo heterozygous frameshift pathogenic variant in TBR1, leading to an early premature termination codon (c.26del, p.(Pro9Leufs*12)). CONCLUSION Based on this case report, we discuss the role of TBR1 in general brain development, language development, intellectual disability and other symptoms of ASD. Providing a detailed clinical description of the individuals with such pathogenic variants should help to understand the genotype-phenotype relationships in ASD.
Collapse
Affiliation(s)
- Laurie-Anne Sapey-Triomphe
- Lyon Neuroscience Research Center, Brain Dynamics and Cognition team, INSERM UMRS 1028, CNRS UMR 5292, Université Claude Bernard Lyon 1, Université de Lyon, F-69000, Lyon, France
- Laboratory of Experimental Psychology, Department of Brain and Cognition, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Julie Reversat
- Lyon Hospitals, Genetics Service and National Reference Centre for Developmental Anomalies, Lyon, France
| | - Gaëtan Lesca
- Lyon Hospitals, Genetics Service and National Reference Centre for Developmental Anomalies, Lyon, France
- Lyon Neuroscience Research Center, Genetics of Neurodevelopment team, INSERM UMRS 1028, CNRS UMR 5292, Université Claude Bernard Lyon 1, Université de Lyon, F-69000, Lyon, France
| | - Nicolas Chatron
- Lyon Hospitals, Genetics Service and National Reference Centre for Developmental Anomalies, Lyon, France
- Lyon Neuroscience Research Center, Genetics of Neurodevelopment team, INSERM UMRS 1028, CNRS UMR 5292, Université Claude Bernard Lyon 1, Université de Lyon, F-69000, Lyon, France
| | - Marina Bussa
- Centre de Ressource Autisme Rhône-Alpes, Centre Hospitalier Le Vinatier, Bron, France
- Hôpital Saint-Jean-de-Dieu, Lyon, France
| | - Sylvie Mazoyer
- Lyon Neuroscience Research Center, Genetics of Neurodevelopment team, INSERM UMRS 1028, CNRS UMR 5292, Université Claude Bernard Lyon 1, Université de Lyon, F-69000, Lyon, France
| | - Christina Schmitz
- Lyon Neuroscience Research Center, Brain Dynamics and Cognition team, INSERM UMRS 1028, CNRS UMR 5292, Université Claude Bernard Lyon 1, Université de Lyon, F-69000, Lyon, France.
| | - Sandrine Sonié
- Lyon Neuroscience Research Center, Brain Dynamics and Cognition team, INSERM UMRS 1028, CNRS UMR 5292, Université Claude Bernard Lyon 1, Université de Lyon, F-69000, Lyon, France
- Centre de Ressource Autisme Rhône-Alpes, Centre Hospitalier Le Vinatier, Bron, France
- Hôpital Saint-Jean-de-Dieu, Lyon, France
| | - Patrick Edery
- Lyon Hospitals, Genetics Service and National Reference Centre for Developmental Anomalies, Lyon, France
- Lyon Neuroscience Research Center, Genetics of Neurodevelopment team, INSERM UMRS 1028, CNRS UMR 5292, Université Claude Bernard Lyon 1, Université de Lyon, F-69000, Lyon, France
| |
Collapse
|
16
|
Hashem S, Nisar S, Bhat AA, Yadav SK, Azeem MW, Bagga P, Fakhro K, Reddy R, Frenneaux MP, Haris M. Genetics of structural and functional brain changes in autism spectrum disorder. Transl Psychiatry 2020; 10:229. [PMID: 32661244 PMCID: PMC7359361 DOI: 10.1038/s41398-020-00921-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/05/2020] [Accepted: 06/09/2020] [Indexed: 12/21/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurological and developmental disorder characterized by social impairment and restricted interactive and communicative behaviors. It may occur as an isolated disorder or in the context of other neurological, psychiatric, developmental, and genetic disorders. Due to rapid developments in genomics and imaging technologies, imaging genetics studies of ASD have evolved in the last few years. Increased risk for ASD diagnosis is found to be related to many specific single-nucleotide polymorphisms, and the study of genetic mechanisms and noninvasive imaging has opened various approaches that can help diagnose ASD at the nascent level. Identifying risk genes related to structural and functional changes in the brain of ASD patients provide a better understanding of the disease's neuropsychiatry and can help identify targets for therapeutic intervention that could be useful for the clinical management of ASD patients.
Collapse
Affiliation(s)
- Sheema Hashem
- Functional and Molecular Imaging Laboratory, Sidra Medicine, Doha, Qatar
| | - Sabah Nisar
- Functional and Molecular Imaging Laboratory, Sidra Medicine, Doha, Qatar
| | - Ajaz A Bhat
- Functional and Molecular Imaging Laboratory, Sidra Medicine, Doha, Qatar
| | | | | | - Puneet Bagga
- Center for Magnetic Resonance and Optical Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Khalid Fakhro
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
- Department of Genetic Medicine, Weill Cornell Medical College, Doha, Qatar
| | - Ravinder Reddy
- Center for Magnetic Resonance and Optical Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | - Mohammad Haris
- Functional and Molecular Imaging Laboratory, Sidra Medicine, Doha, Qatar.
- Laboratory Animal Research Center, Qatar University, Doha, Qatar.
| |
Collapse
|
17
|
Shrestha A, Sultana R, Adeniyi PA, Lee CC, Ogundele OM. Positive Modulation of SK Channel Impedes Neuron-Specific Cytoskeletal Organization and Maturation. Dev Neurosci 2020; 42:59-71. [PMID: 32580196 PMCID: PMC7486235 DOI: 10.1159/000507989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 04/15/2020] [Indexed: 01/01/2023] Open
Abstract
N-methyl-D-aspartate receptor (NMDAR) modulates the structural plasticity of dendritic spines by impacting cytoskeletal organization and kinase signaling. In the developing nervous system, activation of NMDAR is pertinent for neuronal migration, neurite differentiation, and cellular organization. Given that small conductance potassium channels (SK2/3) repress NMDAR ionotropic signaling, this study highlights the impact of neonatal SK channel potentiation on adult cortical and hippocampal organization. Neonatal SK channel potentiation was performed by one injection of SK2/3 agonist (CyPPA) into the pallium of mice on postnatal day 2 (P2). When the animals reached adulthood (P55), the hippocampus and cortex were examined to assess neuronal maturation, lamination, and the distribution of synaptic cytoskeletal proteins. Immunodetection of neuronal markers in the brain of P2-treated P55 mice revealed the presence of immature neurons in the upper cortical layers (layers II-IV) and CA1 (hippocampus). Also, layer-dependent cortical-cell density was attenuated due to the ectopic localization of mature (NeuN+) and immature (Doublecortin+ [DCX+]) neurons in cortical layers II-IV. Similarly, the decreased count of NeuN+ neurons in the CA1 is accompanied by an increase in the number of immature DCX+ neurons. Ectopic localization of neurons in the upper cortex and CA1 caused the dramatic expression of neuron-specific cytoskeletal proteins. In line with this, structural deformity of neuronal projections and the loss of postsynaptic densities suggests that postsynaptic integrity is compromised in the SK2/3+ brain. From these results, we deduced that SK channel activity in the developing brain likely impacts neuronal maturation through its effects on cytoskeletal formation.
Collapse
Affiliation(s)
- Amita Shrestha
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Razia Sultana
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Philip A Adeniyi
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Charles C Lee
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Olalekan M Ogundele
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA,
| |
Collapse
|
18
|
Serth J, Peters I, Dubrowinskaja N, Reese C, Albrecht K, Klintschar M, Lafos M, Grote A, Becker A, Hennenlotter J, Stenzl A, Tezval H, Kuczyk MA. Age-, tumor-, and metastatic tissue-associated DNA hypermethylation of a T-box brain 1 locus in human kidney tissue. Clin Epigenetics 2020; 12:33. [PMID: 32070431 PMCID: PMC7029553 DOI: 10.1186/s13148-020-0823-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/03/2020] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND While a considerable number of tumor-specific hypermethylated loci have been identified in renal cell cancer (RCC), DNA methylation of loci showing successive increases in normal, tumoral, and metastatic tissues could point to genes with high relevance both for the process of tumor development and progression. Here, we report that DNA methylation of a locus in a genomic region corresponding to the 3'UTR of the transcription factor T-box brain 1 (TBR1) mRNA accumulates in normal renal tissues with age and possibly increased body mass index. Moreover, a further tissue-specific increase of methylation was observed for tumor and metastatic tissue samples. RESULTS Biometric analyses of the TCGA KIRC methylation data revealed candidate loci for age-dependent and tumor-specific DNA methylation within the last exon and in a genomic region corresponding to the 3'UTR TBR1 mRNA. To evaluate whether methylation of TBR1 shows association with RCC carcinogenesis, we measured 15 tumor cell lines and 907 renal tissue samples including 355 normal tissues, 175 tissue pairs of normal tumor adjacent and corresponding tumor tissue as well 202 metastatic tissues samples of lung, bone, and brain metastases by the use of pyrosequencing. Statistical evaluation demonstrated age-dependent methylation in normal tissue (R = 0.72, p < 2 × 10-16), association with adiposity (P = 0.019) and tumor-specific hypermethylation (P = 6.1 × 10-19) for RCC tissues. Comparison of tumor and metastatic tissues revealed higher methylation in renal cancer metastases (P = 2.65 × 10-6). CONCLUSIONS Our analyses provide statistical evidence of association between methylation of TBR1 and RCC development and disease progression.
Collapse
Affiliation(s)
- Jürgen Serth
- Klinik für Urologie und urologische Onkologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, D-30625, Hannover, Germany.
| | - Inga Peters
- Klinik für Urologie und urologische Onkologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, D-30625, Hannover, Germany
| | - Natalia Dubrowinskaja
- Klinik für Urologie und urologische Onkologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, D-30625, Hannover, Germany
| | - Christel Reese
- Klinik für Urologie und urologische Onkologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, D-30625, Hannover, Germany
| | - Knut Albrecht
- Brandenburgisches Landesinstitut für Rechtsmedizin, Lindstedter Chaussee 6, D-14469, Potsdam, Germany
| | - Michael Klintschar
- Institut für Rechtsmedizin, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, D-30625, Hannover, Germany
| | - Marcel Lafos
- Institut für Pathologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, D-30625, Hannover, Germany
| | - Alexander Grote
- Evangelisches Klinikum Bethel, Klinik für Neurochirurgie, Burgsteig 13, D-33617, Bielefeld, Germany
| | - Albert Becker
- Institut für Neuropathologie, Universitätsklinikum Bonn, Sigmund Freud Str. 25, D-53105, Bonn, Germany
| | - Jörg Hennenlotter
- Universitätsklinikum Tübingen, Klinik für Urologie, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany
| | - Arnulf Stenzl
- Universitätsklinikum Tübingen, Klinik für Urologie, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany
| | - Hossein Tezval
- Klinik für Urologie und urologische Onkologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, D-30625, Hannover, Germany
| | - Markus A Kuczyk
- Klinik für Urologie und urologische Onkologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, D-30625, Hannover, Germany
| |
Collapse
|
19
|
Artificial Intelligence and the detection of pediatric concussion using epigenomic analysis. Brain Res 2019; 1726:146510. [PMID: 31628932 DOI: 10.1016/j.brainres.2019.146510] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 12/12/2022]
Abstract
Concussion, also referred to as mild traumatic brain injury (mTBI) is the most common type of traumatic brain injury. Currently concussion is an area ofintensescientific interest to better understand the biological mechanisms and for biomarker development. We evaluated whole genome-wide blood DNA cytosine ('CpG') methylation in 17 pediatric concussion isolated cases and 18 unaffected controls using Illumina Infinium MethylationEPIC assay. Pathway analysis was performed using Ingenuity Pathway Analysis to help elucidate the epigenetic and molecular mechanisms of the disorder. Area under the receiver operating characteristics (AUC) curves and FDR p-values were calculated for mTBI detection based on CpG methylation levels. Multiple Artificial Intelligence (AI) platforms including Deep Learning (DL), the newest form of AI, were used to predict concussion based on i) CpG methylation markers alone, and ii) combined epigenetic, clinical and demographic predictors. We found 449 CpG sites (473 genes), those were statistically significantly methylated in mTBI compared to controls. There were four CpGs with excellent individual accuracy (AUC ≥ 0.90-1.00) while 119 displayed good accuracy (AUC ≥ 0.80-0.89) for the prediction of mTBI. The CpG methylation changes ≥10% were observed in many CpG loci after concussion suggesting biological significance. Pathway analysis identified several biologically important neurological pathways that were perturbed including those associated with: impaired brain function, cognition, memory, neurotransmission, intellectual disability and behavioral change and associated disorders. The combination of epigenomic and clinical predictors were highly accurate for the detection of concusion using AI techniques. Using DL/AI, a combination of epigenomic and clinical markers had sensitivity and specificity ≧95% for prediction of mTBI. In this novel study, we identified significant methylation changes in multiple genes in response to mTBI. Gene pathways that were epigenetically dysregulated included several known to be involved in neurological function, thus giving biological plausibility to our findings.
Collapse
|
20
|
Yook C, Kim K, Kim D, Kang H, Kim SG, Kim E, Kim SY. A TBR1-K228E Mutation Induces Tbr1 Upregulation, Altered Cortical Distribution of Interneurons, Increased Inhibitory Synaptic Transmission, and Autistic-Like Behavioral Deficits in Mice. Front Mol Neurosci 2019; 12:241. [PMID: 31680851 PMCID: PMC6797848 DOI: 10.3389/fnmol.2019.00241] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 09/20/2019] [Indexed: 12/11/2022] Open
Abstract
Mutations in Tbr1, a high-confidence ASD (autism spectrum disorder)-risk gene encoding the transcriptional regulator TBR1, have been shown to induce diverse ASD-related molecular, synaptic, neuronal, and behavioral dysfunctions in mice. However, whether Tbr1 mutations derived from autistic individuals cause similar dysfunctions in mice remains unclear. Here we generated and characterized mice carrying the TBR1-K228E de novo mutation identified in human ASD and identified various ASD-related phenotypes. In heterozygous mice carrying this mutation (Tbr1+/K228E mice), levels of the TBR1-K228E protein, which is unable to bind target DNA, were strongly increased. RNA-Seq analysis of the Tbr1+/K228E embryonic brain indicated significant changes in the expression of genes associated with neurons, astrocytes, ribosomes, neuronal synapses, and ASD risk. The Tbr1+/K228E neocortex also displayed an abnormal distribution of parvalbumin-positive interneurons, with a lower density in superficial layers but a higher density in deep layers. These changes were associated with an increase in inhibitory synaptic transmission in layer 6 pyramidal neurons that was resistant to compensation by network activity. Behaviorally, Tbr1+/K228E mice showed decreased social interaction, increased self-grooming, and modestly increased anxiety-like behaviors. These results suggest that the human heterozygous TBR1-K228E mutation induces ASD-related transcriptomic, protein, neuronal, synaptic, and behavioral dysfunctions in mice.
Collapse
Affiliation(s)
- Chaehyun Yook
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, South Korea
| | - Kyungdeok Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, South Korea
| | - Doyoun Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information (KISTI), Daejeon, South Korea
| | - Sun-Gyun Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, South Korea.,Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Soo Young Kim
- College of Pharmacy, Yeongnam University, Gyeongsan, South Korea
| |
Collapse
|
21
|
Lau YFC, Li Y, Kido T. Battle of the sexes: contrasting roles of testis-specific protein Y-encoded (TSPY) and TSPX in human oncogenesis. Asian J Androl 2019; 21:260-269. [PMID: 29974883 PMCID: PMC6498724 DOI: 10.4103/aja.aja_43_18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/17/2018] [Indexed: 12/13/2022] Open
Abstract
The Y-located testis-specific protein Y-encoded (TSPY) and its X-homologue TSPX originated from the same ancestral gene, but act as a proto-oncogene and a tumor suppressor gene, respectively. TSPY has specialized in male-specific functions, while TSPX has assumed the functions of the ancestral gene. Both TSPY and TSPX harbor a conserved SET/NAP domain, but are divergent at flanking structures. Specifically, TSPX contains a C-terminal acidic domain, absent in TSPY. They possess contrasting properties, in which TSPY and TSPX, respectively, accelerate and arrest cell proliferation, stimulate and inhibit cyclin B-CDK1 phosphorylation activities, have no effect and promote proteosomal degradation of the viral HBx oncoprotein, and exacerbate and repress androgen receptor (AR) and constitutively active AR variant, such as AR-V7, gene transactivation. The inhibitory domain has been mapped to the carboxyl acidic domain in TSPX, truncation of which results in an abbreviated TSPX exerting positive actions as TSPY. Transposition of the acidic domain to the C-terminus of TSPY results in an inhibitory protein as intact TSPX. Hence, genomic mutations/aberrant splicing events could generate TSPX proteins with truncated acidic domain and oncogenic properties as those for TSPY. Further, TSPY is upregulated by AR and AR-V7 in ligand-dependent and ligand-independent manners, respectively, suggesting the existence of a positive feedback loop between a Y-located proto-oncogene and male sex hormone/receptors, thereby amplifying the respective male oncogenic actions in human cancers and diseases. TSPX counteracts such positive feedback loop. Hence, TSPY and TSPX are homologues on the sex chromosomes that function at the two extremes of the human oncogenic spectrum.
Collapse
Affiliation(s)
- Yun-Fai Chris Lau
- Division of Cell and Developmental Genetics, Department of Medicine, VA Medical Center and Institute for Human Genetics, University of California, San Francisco, CA 94121, USA
| | - Yunmin Li
- Division of Cell and Developmental Genetics, Department of Medicine, VA Medical Center and Institute for Human Genetics, University of California, San Francisco, CA 94121, USA
| | - Tatsuo Kido
- Division of Cell and Developmental Genetics, Department of Medicine, VA Medical Center and Institute for Human Genetics, University of California, San Francisco, CA 94121, USA
| |
Collapse
|
22
|
Geisler S, Schöpf CL, Stanika R, Kalb M, Campiglio M, Repetto D, Traxler L, Missler M, Obermair GJ. Presynaptic α 2δ-2 Calcium Channel Subunits Regulate Postsynaptic GABA A Receptor Abundance and Axonal Wiring. J Neurosci 2019; 39:2581-2605. [PMID: 30683685 PMCID: PMC6445987 DOI: 10.1523/jneurosci.2234-18.2019] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 01/03/2019] [Accepted: 01/08/2019] [Indexed: 01/26/2023] Open
Abstract
Presynaptic α2δ subunits of voltage-gated calcium channels regulate channel abundance and are involved in glutamatergic synapse formation. However, little is known about the specific functions of the individual α2δ isoforms and their role in GABAergic synapses. Using primary neuronal cultures of embryonic mice of both sexes, we here report that presynaptic overexpression of α2δ-2 in GABAergic synapses strongly increases clustering of postsynaptic GABAARs. Strikingly, presynaptic α2δ-2 exerts the same effect in glutamatergic synapses, leading to a mismatched localization of GABAARs. This mismatching is caused by an aberrant wiring of glutamatergic presynaptic boutons with GABAergic postsynaptic positions. The trans-synaptic effect of α2δ-2 is independent of the prototypical cell-adhesion molecules α-neurexins (α-Nrxns); however, α-Nrxns together with α2δ-2 can modulate postsynaptic GABAAR abundance. Finally, exclusion of the alternatively spliced exon 23 of α2δ-2 is essential for the trans-synaptic mechanism. The novel function of α2δ-2 identified here may explain how abnormal α2δ subunit expression can cause excitatory-inhibitory imbalance often associated with neuropsychiatric disorders.SIGNIFICANCE STATEMENT Voltage-gated calcium channels regulate important neuronal functions such as synaptic transmission. α2δ subunits modulate calcium channels and are emerging as regulators of brain connectivity. However, little is known about how individual α2δ subunits contribute to synapse specificity. Here, we show that presynaptic expression of a single α2δ variant can modulate synaptic connectivity and the localization of inhibitory postsynaptic receptors. Our findings provide basic insights into the development of specific synaptic connections between nerve cells and contribute to our understanding of normal nerve cell functions. Furthermore, the identified mechanism may explain how an altered expression of calcium channel subunits can result in aberrant neuronal wiring often associated with neuropsychiatric disorders such as autism or schizophrenia.
Collapse
Affiliation(s)
- Stefanie Geisler
- Division of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria, and
| | - Clemens L Schöpf
- Division of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria, and
| | - Ruslan Stanika
- Division of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria, and
| | - Marcus Kalb
- Division of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria, and
| | - Marta Campiglio
- Division of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria, and
| | - Daniele Repetto
- Institute of Anatomy and Molecular Neurobiology, Westfälische Wilhelms-University, 48149 Münster, Germany
| | - Larissa Traxler
- Division of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria, and
| | - Markus Missler
- Institute of Anatomy and Molecular Neurobiology, Westfälische Wilhelms-University, 48149 Münster, Germany
| | - Gerald J Obermair
- Division of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria, and
| |
Collapse
|
23
|
The primate-specific peptide Y-P30 regulates morphological maturation of neocortical dendritic spines. PLoS One 2019; 14:e0211151. [PMID: 30759095 PMCID: PMC6373909 DOI: 10.1371/journal.pone.0211151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/08/2019] [Indexed: 12/20/2022] Open
Abstract
The 30-amino acid peptide Y-P30 corresponds to the N-terminus of the primate-specific, sweat gland-derived dermcidin prepropeptide. Previous work has revealed that Y-P30 enhances the interaction of pleiotrophin and syndecans-2/3, and thus represents a natural ligand to study this signaling pathway. In immature neurons, Y-P30 activates the c-Src and p42/44 ERK kinase pathway, increases the amount of F-actin in axonal growth cones, and promotes neuronal survival, cell migration and axonal elongation. The action of Y-P30 on axonal growth requires syndecan-3 and heparan sulfate side chains. Whether Y-P30 has the potential to influence dendrites and dendritic protrusions has not been explored. The latter is suggested by the observations that syndecan-2 expression increases during postnatal development, that syndecan-2 becomes enriched in dendritic spines, and that overexpression of syndecan-2 in immature neurons results in a premature morphological maturation of dendritic spines. Here, analysing rat cortical pyramidal and non-pyramidal neurons in organotypic cultures, we show that Y-P30 does not alter the development of the dendritic arborization patterns. However, Y-P30 treatment decreases the density of apical, but not basal dendritic protrusions at the expense of the filopodia. Analysis of spine morphology revealed an unchanged mushroom/stubby-to-thin spine ratio and a shortening of the longest decile of dendritic protrusions. Whole-cell recordings from cortical principal neurons in dissociated cultures grown in the presence of Y-P30 demonstrated a decrease in the frequency of glutamatergic mEPSCs. Despite these differences in protrusion morphology and synaptic transmission, the latter likely attributable to presynaptic effects, calcium event rate and amplitude recorded in pyramidal neurons in organotypic cultures were not altered by Y-P30 treatment. Together, our data suggest that Y-P30 has the capacity to decelerate spinogenesis and to promote morphological, but not synaptic, maturation of dendritic protrusions.
Collapse
|
24
|
P2X7 Receptors Drive Poly(I:C) Induced Autism-like Behavior in Mice. J Neurosci 2019; 39:2542-2561. [PMID: 30683682 DOI: 10.1523/jneurosci.1895-18.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 01/09/2019] [Accepted: 01/12/2019] [Indexed: 02/08/2023] Open
Abstract
Maternal immune activation (MIA) is a principal environmental risk factor contributing to autism spectrum disorder (ASD), which compromises fetal brain development at critical periods of pregnancy and might be causally linked to ASD symptoms. We report that endogenous activation of the purinergic ion channel P2X7 (P2rx7) is necessary and sufficient to transduce MIA to autistic phenotype in male offspring. MIA induced by poly(I:C) injections to P2rx7 WT mouse dams elicited an autism-like phenotype in their offspring, and these alterations were not observed in P2rx7-deficient mice, or following maternal treatment with a specific P2rx7 antagonist, JNJ47965567. Genetic deletion and pharmacological inhibition of maternal P2rx7s also counteracted the induction of IL-6 in the maternal plasma and fetal brain, and disrupted brain development, whereas postnatal P2rx7 inhibition alleviated behavioral and morphological alterations in the offspring. Administration of ATP to P2rx7 WT dams also evoked autistic phenotype, but not in KO dams, implying that P2rx7 activation by ATP is sufficient to induce autism-like features in offspring. Our results point to maternal and offspring P2rx7s as potential therapeutic targets for the early prevention and treatment of ASD.SIGNIFICANCE STATEMENT Autism spectrum disorder (ASD) is a neurodevelopmental psychiatric disorder caused by genetic and environmental factors. Recent studies highlighted the importance of perinatal risks, in particular, maternal immune activation (MIA), showing strong association with the later emergence of ASD in the affected children. MIA could be mimicked in animal models via injection of a nonpathogenic agent poly(I:C) during pregnancy. This is the first report showing the key role of a ligand gated ion channel, the purinergic P2X7 receptor in MIA-induced autism-like behavioral and biochemical features. We show that genetic or pharmacological inhibition of both maternal and offspring P2X7 receptors could reverse the compromised brain development and autistic phenotype pointing to new possibilities for prevention and treatment of ASD.
Collapse
|
25
|
Mori T, Kasem EA, Suzuki-Kouyama E, Cao X, Li X, Kurihara T, Uemura T, Yanagawa T, Tabuchi K. Deficiency of calcium/calmodulin-dependent serine protein kinase disrupts the excitatory-inhibitory balance of synapses by down-regulating GluN2B. Mol Psychiatry 2019; 24:1079-1092. [PMID: 30610199 PMCID: PMC6756202 DOI: 10.1038/s41380-018-0338-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 11/27/2018] [Accepted: 12/10/2018] [Indexed: 11/24/2022]
Abstract
Calcium/calmodulin-dependent serine protein kinase (CASK) is a membrane-associated guanylate kinase (MAGUK) protein that is associated with neurodevelopmental disorders. CASK is thought to have both pre- and postsynaptic functions, but the mechanism and consequences of its functions in the brain have yet to be elucidated, because homozygous CASK-knockout (CASK-KO) mice die before brain maturation. Taking advantage of the X-chromosome inactivation (XCI) mechanism, here we examined the synaptic functions of CASK-KO neurons in acute brain slices of heterozygous CASK-KO female mice. We also analyzed CASK-knockdown (KD) neurons in acute brain slices generated by in utero electroporation. Both CASK-KO and CASK-KD neurons showed a disruption of the excitatory and inhibitory (E/I) balance. We further found that the expression level of the N-methyl-D-aspartate receptor subunit GluN2B was decreased in CASK-KD neurons and that overexpressing GluN2B rescued the disrupted E/I balance in CASK-KD neurons. These results suggest that the down-regulation of GluN2B may be involved in the mechanism of the disruption of synaptic E/I balance in CASK-deficient neurons.
Collapse
Affiliation(s)
- Takuma Mori
- 0000 0001 1507 4692grid.263518.bDepartment of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621 Japan
| | - Enas A. Kasem
- 0000 0001 1507 4692grid.263518.bDepartment of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621 Japan ,0000 0004 0578 3577grid.411978.2Department of Zoology, Faculty of Science, Kafr Elsheikh University, Kafr Elsheihk, 33511 Egypt
| | - Emi Suzuki-Kouyama
- 0000 0001 1507 4692grid.263518.bDepartment of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621 Japan
| | - Xueshan Cao
- 0000 0001 1507 4692grid.263518.bDepartment of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621 Japan
| | - Xue Li
- 0000 0001 1507 4692grid.263518.bDepartment of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621 Japan
| | - Taiga Kurihara
- 0000 0001 1507 4692grid.263518.bDepartment of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621 Japan
| | - Takeshi Uemura
- 0000 0001 1507 4692grid.263518.bDepartment of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621 Japan ,0000 0001 1507 4692grid.263518.bInstitute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, 390-8621 Japan ,0000 0004 1754 9200grid.419082.6CREST, JST, Saitama, 332-0012 Japan
| | - Toru Yanagawa
- 0000 0001 2369 4728grid.20515.33Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575 Japan
| | - Katsuhiko Tabuchi
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621, Japan. .,Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, 390-8621, Japan. .,PRESTO, JST, Saitama, 332-0012, Japan.
| |
Collapse
|
26
|
Chen YC, Chang YW, Huang YS. Dysregulated Translation in Neurodevelopmental Disorders: An Overview of Autism-Risk Genes Involved in Translation. Dev Neurobiol 2018; 79:60-74. [PMID: 30430754 DOI: 10.1002/dneu.22653] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 09/17/2018] [Accepted: 10/25/2018] [Indexed: 01/08/2023]
Abstract
Regulated local translation-whereby specific mRNAs are transported and localized in subcellular domains where they are translated in response to regional signals-allows for remote control of gene expression to concentrate proteins in subcellular compartments. Neurons are highly polarized cells with unique features favoring local control for axonal pathfinding and synaptic plasticity, which are key processes involved in constructing functional circuits in the developing brain. Neurodevelopmental disorders are caused by genetic or environmental factors that disturb the nervous system's development during prenatal and early childhood periods. The growing list of genetic mutations that affect mRNA translation raises the question of whether aberrant translatomes in individuals with neurodevelopmental disorders share common molecular features underlying their stereotypical phenotypes and, vice versa, cause a certain degree of phenotypic heterogeneity. Here, we briefly give an overview of the role of local translation during neuronal development. We take the autism-risk gene list and discuss the molecules that (perhaps) are involved in mRNA transport and translation. Both exaggerated and suppressed translation caused by mutations in those genes have been identified or suggested. Finally, we discuss some proof-of-principle regimens for use in autism mouse models to correct dysregulated translation.
Collapse
Affiliation(s)
- Yan-Chu Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Yu-Wei Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Yi-Shuian Huang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| |
Collapse
|
27
|
Maternal high fat diet alters offspring epigenetic regulators, amygdala glutamatergic profile and anxiety. Psychoneuroendocrinology 2018; 96:132-141. [PMID: 29940426 DOI: 10.1016/j.psyneuen.2018.06.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/12/2018] [Accepted: 06/18/2018] [Indexed: 12/31/2022]
Abstract
Maternal obesity during pregnancy can impact long-term health, predisposition to disease, and risk of neurological disorders in offspring. This may arise from disruption to epigenetic processes during offspring brain development. Using a maternal high fat diet (mHFD) mouse model, we investigated the expression of genes encoding epigenetic regulators in the brains of gestational day (GD) 17.5 mHFD offspring. We found significant, regionally unique changes in expression of epigenetic regulators in the developing brain of mHFD offspring compared to controls, with Gadd45b downregulated in medial prefrontal cortex, Mecp2 downregulated in amygdala, and sex-specific downregulation of Crebbp, Dnmt3b, and Mecp2 in male mHFD hippocampus. Decreased Mecp2 in the amygdala was associated with significant upregulation of the Mecp2-repressed gene, Tbr1, and an increased number of TBR1+ glutamatergic neurons in the basomedial nucleus of the amygdala. Tbr1 upregulation in amygdala was also observed in postnatal day 8 (P8) mHFD offspring, and levels of glutamate receptor gene Grin2b, and Fos, a marker for neuronal activity, were increased. Indications of heightened excitatory drive in mHFD offspring amygdala were associated with an anxiety-like phenotype, with mHFD offspring displaying altered ultrasonic vocalization characteristics at P8, and adult female mHFD offspring spending decreased time on the open arm of the Elevated Plus Maze. Together, this data provides insight into sex-specific offspring vulnerability to perinatal mHFD programming of anxiety-like behaviors.
Collapse
|
28
|
Del Tufo SN, Frost SJ, Hoeft F, Cutting LE, Molfese PJ, Mason GF, Rothman DL, Fulbright RK, Pugh KR. Neurochemistry Predicts Convergence of Written and Spoken Language: A Proton Magnetic Resonance Spectroscopy Study of Cross-Modal Language Integration. Front Psychol 2018; 9:1507. [PMID: 30233445 PMCID: PMC6131664 DOI: 10.3389/fpsyg.2018.01507] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 07/30/2018] [Indexed: 12/26/2022] Open
Abstract
Recent studies have provided evidence of associations between neurochemistry and reading (dis)ability (Pugh et al., 2014). Based on a long history of studies indicating that fluent reading entails the automatic convergence of the written and spoken forms of language and our recently proposed Neural Noise Hypothesis (Hancock et al., 2017), we hypothesized that individual differences in cross-modal integration would mediate, at least partially, the relationship between neurochemical concentrations and reading. Cross-modal integration was measured in 231 children using a two-alternative forced choice cross-modal matching task with three language conditions (letters, words, and pseudowords) and two levels of difficulty within each language condition. Neurometabolite concentrations of Choline (Cho), Glutamate (Glu), gamma-Aminobutyric (GABA), and N- acetyl-aspartate (NAA) were then measured in a subset of this sample (n = 70) with Magnetic Resonance Spectroscopy (MRS). A structural equation mediation model revealed that the effect of cross-modal word matching mediated the relationship between increased Glu (which has been proposed to be an index of neural noise) and poorer reading ability. In addition, the effect of cross-modal word matching fully mediated a relationship between increased Cho and poorer reading ability. Multilevel mixed effects models confirmed that lower Cho predicted faster cross-modal matching reaction time, specifically in the hard word condition. These Cho findings are consistent with previous work in both adults and children showing a negative association between Cho and reading ability. We also found two novel neurochemical relationships. Specifically, lower GABA and higher NAA predicted faster cross-modal matching reaction times. We interpret these results within a biochemical framework in which the ability of neurochemistry to predict reading ability may at least partially be explained by cross-modal integration.
Collapse
Affiliation(s)
- Stephanie N Del Tufo
- Department of Special Education, Peabody College, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, United States.,Haskins Laboratories, New Haven, CT, United States
| | | | - Fumiko Hoeft
- Haskins Laboratories, New Haven, CT, United States.,Department of Psychiatry, University of California, San Francisco, San Francisco, CA, United States
| | - Laurie E Cutting
- Department of Special Education, Peabody College, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, United States.,Haskins Laboratories, New Haven, CT, United States.,Peabody College of Education and Human Development, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN, United States
| | - Peter J Molfese
- Haskins Laboratories, New Haven, CT, United States.,Section on Functional Imaging Methods, Laboratory of Brain and Cognition, Department of Health and Human Services, National Institutes of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Graeme F Mason
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States.,Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Douglas L Rothman
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States.,Department of Biomedical Engineering, Yale University School of Medicine, New Haven, CT, United States
| | - Robert K Fulbright
- Haskins Laboratories, New Haven, CT, United States.,Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States
| | - Kenneth R Pugh
- Haskins Laboratories, New Haven, CT, United States.,Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States.,Department of Psychological Sciences, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
29
|
Liu J, Reggiani JDS, Laboulaye MA, Pandey S, Chen B, Rubenstein JLR, Krishnaswamy A, Sanes JR. Tbr1 instructs laminar patterning of retinal ganglion cell dendrites. Nat Neurosci 2018; 21:659-670. [PMID: 29632360 PMCID: PMC5920715 DOI: 10.1038/s41593-018-0127-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/20/2018] [Indexed: 12/20/2022]
Abstract
Visual information is delivered to the brain by >40 types of retinal ganglion cells (RGCs). Diversity in this representation arises within the inner plexiform layer (IPL), where dendrites of each RGC type are restricted to specific sublaminae, limiting the interneuronal types that can innervate them. How such dendritic restriction arises is unclear. We show that the transcription factor Tbr1 is expressed by four mouse RGC types with dendrites in the outer IPL and is required for their laminar specification. Loss of Tbr1 results in elaboration of dendrites within the inner IPL, while misexpression in other cells retargets their neurites to the outer IPL. Two transmembrane molecules, Sorcs3 and Cdh8, act as effectors of the Tbr1-controlled lamination program. However, they are expressed in just one Tbr1+ RGC type, supporting a model in which a single transcription factor implements similar laminar choices in distinct cell types by recruiting partially non-overlapping effectors.
Collapse
Affiliation(s)
- Jinyue Liu
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA.,Center for Brain Science, Harvard University, Cambridge, MA, USA.,Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | - Jasmine D S Reggiani
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA.,Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Mallory A Laboulaye
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA.,Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Shristi Pandey
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA.,Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Bin Chen
- Department of Molecular, Cell and Developmental Biology, University of California at Santa Cruz, Santa Cruz, CA, USA
| | - John L R Rubenstein
- Department of Psychiatry, University of California at San Francisco, San Francisco, CA, USA
| | - Arjun Krishnaswamy
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA.,Center for Brain Science, Harvard University, Cambridge, MA, USA.,Department of Physiology, McGill University, Montreal, QC, Canada
| | - Joshua R Sanes
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA. .,Center for Brain Science, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
30
|
Adorjan I, Ahmed B, Feher V, Torso M, Krug K, Esiri M, Chance SA, Szele FG. Calretinin interneuron density in the caudate nucleus is lower in autism spectrum disorder. Brain 2017; 140:2028-2040. [PMID: 29177493 DOI: 10.1093/brain/awx131] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 04/18/2017] [Indexed: 02/05/2023] Open
Abstract
Autism spectrum disorder is a debilitating condition with possible neurodevelopmental origins but unknown neuroanatomical correlates. Whereas investigators have paid much attention to the cerebral cortex, few studies have detailed the basal ganglia in autism. The caudate nucleus may be involved in the repetitive movements and limbic changes of autism. We used immunohistochemistry for calretinin and neuropeptide Y in 24 age- and gender-matched patients with autism spectrum disorder and control subjects ranging in age from 13 to 69 years. Patients with autism had a 35% lower density of calretinin+ interneurons in the caudate that was driven by loss of small calretinin+ neurons. This was not caused by altered size of the caudate, as its cross-sectional surface areas were similar between diagnostic groups. Controls exhibited an age-dependent increase in the density of medium and large calretinin+ neurons, whereas subjects with autism did not. Diagnostic groups did not differ regarding ionized calcium-binding adapter molecule 1+ immunoreactivity for microglia, suggesting chronic inflammation did not cause the decreased calretinin+ density. There was no statistically significant difference in the density of neuropeptide Y+ neurons between subjects with autism and controls. The decreased calretinin+ density may disrupt the excitation/inhibition balance in the caudate leading to dysfunctional corticostriatal circuits. The description of such changes in autism spectrum disorder may clarify pathomechanisms and thereby help identify targets for drug intervention and novel therapeutic strategies.
Collapse
Affiliation(s)
- Istvan Adorjan
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.,Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Bashir Ahmed
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Virginia Feher
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Mario Torso
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Kristine Krug
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Margaret Esiri
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Steven A Chance
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Francis G Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
31
|
Gray LT, Yao Z, Nguyen TN, Kim TK, Zeng H, Tasic B. Layer-specific chromatin accessibility landscapes reveal regulatory networks in adult mouse visual cortex. eLife 2017; 6. [PMID: 28112643 PMCID: PMC5325622 DOI: 10.7554/elife.21883] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 01/22/2017] [Indexed: 12/20/2022] Open
Abstract
Mammalian cortex is a laminar structure, with each layer composed of a characteristic set of cell types with different morphological, electrophysiological, and connectional properties. Here, we define chromatin accessibility landscapes of major, layer-specific excitatory classes of neurons, and compare them to each other and to inhibitory cortical neurons using the Assay for Transposase-Accessible Chromatin with high-throughput sequencing (ATAC-seq). We identify a large number of layer-specific accessible sites, and significant association with genes that are expressed in specific cortical layers. Integration of these data with layer-specific transcriptomic profiles and transcription factor binding motifs enabled us to construct a regulatory network revealing potential key layer-specific regulators, including Cux1/2, Foxp2, Nfia, Pou3f2, and Rorb. This dataset is a valuable resource for identifying candidate layer-specific cis-regulatory elements in adult mouse cortex.
Collapse
Affiliation(s)
- Lucas T Gray
- Allen Institute for Brain Science, Seattle, United States
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, United States
| | | | - Tae Kyung Kim
- Allen Institute for Brain Science, Seattle, United States
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, United States
| | - Bosiljka Tasic
- Allen Institute for Brain Science, Seattle, United States
| |
Collapse
|
32
|
Huang TN, Hsueh YP. Calcium/calmodulin-dependent serine protein kinase (CASK), a protein implicated in mental retardation and autism-spectrum disorders, interacts with T-Brain-1 (TBR1) to control extinction of associative memory in male mice. J Psychiatry Neurosci 2017; 42:37-47. [PMID: 28234597 PMCID: PMC5373711 DOI: 10.1503/jpn.150359] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Human genetic studies have indicated that mutations in calcium/calmodulin-dependent serine protein kinase (CASK) result in X-linked mental retardation and autism-spectrum disorders. We aimed to establish a mouse model to study how Cask regulates mental ability. METHODS Because Cask encodes a multidomain scaffold protein, a possible strategy to dissect how CASK regulates mental ability and cognition is to disrupt specific protein-protein interactions of CASK in vivo and then investigate the impact of individual specific protein interactions. Previous in vitro analyses indicated that a rat CASK T724A mutation reduces the interaction between CASK and T-brain-1 (TBR1) in transfected COS cells. Because TBR1 is critical for glutamate receptor, ionotropic, N-methyl-D-aspartate receptor subunit 2B (Grin2b) expression and is a causative gene for autism and intellectual disability, we then generated CASK T740A (corresponding to rat CASK T724A) mutant mice using a gene-targeting approach. Immunoblotting, coimmunoprecipitation, histological methods and behavioural assays (including home cage, open field, auditory and contextual fear conditioning and conditioned taste aversion) were applied to investigate expression of CASK and its related proteins, the protein-protein interactions of CASK, and anatomic and behavioural features of CASK T740A mice. RESULTS The CASK T740A mutation attenuated the interaction between CASK and TBR1 in the brain. However, CASK T740A mice were generally healthy, without obvious defects in brain morphology. The most dramatic defect among the mutant mice was in extinction of associative memory, though acquisition was normal. LIMITATIONS The functions of other CASK protein interactions cannot be addressed using CASK T740A mice. CONCLUSION Disruption of the CASK and TBR1 interaction impairs extinction, suggesting the involvement of CASK in cognitive flexibility.
Collapse
Affiliation(s)
| | - Yi-Ping Hsueh
- Correspondence to: Y.P. Hsueh, Institute of Molecular Biology, Academia Sinica, 128, Academia Rd., Sec. 2, Taipei, 11529, Taiwan;
| |
Collapse
|
33
|
de Campo DM, Cameron JL, Miano JM, Lewis DA, Mirnics K, Fudge JL. Maternal deprivation alters expression of neural maturation gene tbr1 in the amygdala paralaminar nucleus in infant female macaques. Dev Psychobiol 2016; 59:235-249. [PMID: 27917473 DOI: 10.1002/dev.21493] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/17/2016] [Indexed: 12/12/2022]
Abstract
Early parental loss is associated with social-emotional dysregulation and amygdala physiologic changes. Previously, we examined whole amygdala gene expression in infant monkeys exposed to early maternal deprivation. Here, we focus on an amygdala region with immature neurons at birth: the paralaminar nucleus (PL). We hypothesized that 1) the normal infant PL is enriched in a subset of neural maturation (NM) genes compared to a nearby amygdala subregion; and 2) maternal deprivation would downregulate expression of NM transcripts (mRNA). mRNAs for bcl2, doublecortin, neuroD1, and tbr1-genes expressed in post-mitotic neurons-were enriched in the normal PL. Maternal deprivation at either 1 week or 1 month of age resulted in PL-specific downregulation of tbr1-a transcription factor necessary for directing neuroblasts to a glutamatergic phenotype. tbr1 expression also correlated with typical social behaviors. We conclude that maternal deprivation influences glutamatergic neuronal development in the PL, possibly influencing circuits mediating social learning.
Collapse
Affiliation(s)
- Danielle M de Campo
- Department of Neuroscience, University of Rochester Medical Center, Rochester, New York.,Department of Medicine, University of Rochester Medical Center, Rochester, New York.,Department of Psychiatry, University of Rochester Medical Center, Rochester, New York
| | - Judy L Cameron
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Joseph M Miano
- Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Karoly Mirnics
- Munroe-Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, Nebraska
| | - Julie L Fudge
- Department of Neuroscience, University of Rochester Medical Center, Rochester, New York.,Department of Medicine, University of Rochester Medical Center, Rochester, New York.,Department of Psychiatry, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
34
|
Levetiracetam mitigates doxorubicin-induced DNA and synaptic damage in neurons. Sci Rep 2016; 6:25705. [PMID: 27168474 PMCID: PMC4863375 DOI: 10.1038/srep25705] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 04/21/2016] [Indexed: 02/07/2023] Open
Abstract
Neurotoxicity may occur in cancer patients and survivors during or after chemotherapy. Cognitive deficits associated with neurotoxicity can be subtle or disabling and frequently include disturbances in memory, attention, executive function and processing speed. Searching for pathways altered by anti-cancer treatments in cultured primary neurons, we discovered that doxorubicin, a commonly used anti-neoplastic drug, significantly decreased neuronal survival. The drug promoted the formation of DNA double-strand breaks in primary neurons and reduced synaptic and neurite density. Pretreatment of neurons with levetiracetam, an FDA-approved anti-epileptic drug, enhanced survival of chemotherapy drug-treated neurons, reduced doxorubicin-induced formation of DNA double-strand breaks, and mitigated synaptic and neurite loss. Thus, levetiracetam might be part of a valuable new approach for mitigating synaptic damage and, perhaps, for treating cognitive disturbances in cancer patients and survivors.
Collapse
|