1
|
Bigelow LJ, Jarvis JH, Fiset C, Pope EK, Le Maistre-Matthys C, Benke TA, Bernard PB. Early life seizures and olfactory communication in rats. Epilepsia 2024; 65:3091-3099. [PMID: 39221936 PMCID: PMC11495993 DOI: 10.1111/epi.18099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE Early life seizures (ELS) are commonly associated with autism spectrum disorder (ASD); however, the exact role of ELS in the pathology is unknown. Prior studies have demonstrated social deficits, a core feature of ASD, following ELS; consequently, alterations in sensory modalities may contribute to the overall social deficits. Considering the speculated contribution of sensory deficit to social communication, we examined the developmental consequences of early postnatal kainic acid (KA)-induced seizures on olfactory preference and neural markers in the olfactory bulb in both male and female Sprague Dawley rats. METHODS KA-induced seizures or saline was administered. Rats were then exposed to a series of biologically relevant scents including male scent, female scent, nest scent, and phenylethylamine during the juvenile period and again during adulthood. Alterations in sensory modalities were expected to be expressed via abnormal preference for certain scents and/or production of abnormal ultrasonic vocalizations in response to scents. The olfactory bulbs were also assessed for the biologically relevant markers glial fibrillary acidic protein (GFAP) and calcium/calmodulin-dependent protein kinase II (CAMKII). RESULTS Our findings resulted in no significant differences in olfactory preference following ELS for juveniles or adults compared to controls. Similarly, there were no differences in GFAP expression or the ratio of phosphorylated CAMKII to CAMKII in either olfactory bulb. Interestingly, despite a lack of treatment differences, different scents were shown to elicit different responses in juvenile rats, yet these differences subsided in adulthood. SIGNIFICANCE Overall, the results of this study suggest that olfaction does not contribute to socialization deficit following ELS within the KA model.
Collapse
Affiliation(s)
- Logan J. Bigelow
- University of Prince Edward Island, Biomedical Sciences, Charlottetown, PE, Canada
| | - Jack H.M. Jarvis
- University of Prince Edward Island, Biomedical Sciences, Charlottetown, PE, Canada
| | - Catherine Fiset
- University of Prince Edward Island, Biomedical Sciences, Charlottetown, PE, Canada
| | - Emily K. Pope
- University of Prince Edward Island, Biomedical Sciences, Charlottetown, PE, Canada
| | | | - Tim A. Benke
- University of Colorado School of Medicine, Department of Pediatrics, Aurora, CO, United States
| | - Paul B. Bernard
- University of Prince Edward Island, Biomedical Sciences, Charlottetown, PE, Canada
| |
Collapse
|
2
|
Obot P, Cibelli A, Pan J, Velíšek L, Velíšková J, Scemes E. Pannexin1 Mediates Early-Life Seizure-Induced Social Behavior Deficits. ASN Neuro 2024; 16:2371164. [PMID: 39024558 PMCID: PMC11262470 DOI: 10.1080/17590914.2024.2371164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/11/2024] [Indexed: 07/20/2024] Open
Abstract
There is a high co-morbidity between childhood epilepsy and autism spectrum disorder (ASD), with age of seizure onset being a critical determinant of behavioral outcomes. The interplay between these comorbidities has been investigated in animal models with results showing that the induction of seizures at early post-natal ages leads to learning and memory deficits and to autistic-like behavior in adulthood. Modifications of the excitation/inhibition (glutamate/GABA, ATP/adenosine) balance that follows early-life seizures (ELS) are thought to be the physiological events that underlie neuropsychiatric and neurodevelopmental disorders. Although alterations in purinergic/adenosinergic signaling have been implicated in seizures and ASD, it is unknown whether the ATP release channels, Pannexin1 (Panx1), contribute to ELS-induced behavior changes. To tackle this question, we used the ELS-kainic acid model in transgenic mice with global and cell type specific deletion of Panx1 to evaluate whether these channels were involved in behavioral deficits that occur later in life. Our studies show that ELS results in Panx1 dependent social behavior deficits and also in poor performance in a spatial memory test that does not involve Panx1. These findings provide support for a link between ELS and adult behavioral deficits. Moreover, we identify neuronal and not astrocyte Panx1 as a potential target to specifically limit astrogliosis and social behavioral deficits resultant from early-life seizures.
Collapse
Affiliation(s)
- Price Obot
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
| | - Antonio Cibelli
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jian Pan
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
| | - Libor Velíšek
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
- Department of Neurology, New York Medical College, Valhalla, New York, USA
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Jana Velíšková
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
- Department of Neurology, New York Medical College, Valhalla, New York, USA
- Department of Obstetrics and Gynecology, New York Medical College, Valhalla, New York, USA
| | - Eliana Scemes
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
| |
Collapse
|
3
|
Winden KD, Pham TT, Teaney NA, Ruiz J, Chen R, Chen C, Sahin M. Increased degradation of FMRP contributes to neuronal hyperexcitability in tuberous sclerosis complex. Cell Rep 2023; 42:112838. [PMID: 37494191 PMCID: PMC10529098 DOI: 10.1016/j.celrep.2023.112838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 05/12/2023] [Accepted: 07/05/2023] [Indexed: 07/28/2023] Open
Abstract
Autism spectrum disorder (ASD) is a highly prevalent neurodevelopmental disorder, but new therapies have been impeded by a lack of understanding of the pathological mechanisms. Tuberous sclerosis complex (TSC) and fragile X syndrome are associated with alterations in the mechanistic target of rapamycin (mTOR) and fragile X messenger ribonucleoprotein 1 (FMRP), which have been implicated in the development of ASD. Previously, we observed that transcripts associated with FMRP were down-regulated in TSC2-deficient neurons. In this study, we find that FMRP turnover is dysregulated in TSC2-deficient rodent primary neurons and human induced pluripotent stem cell (iPSC)-derived neurons and is dependent on the E3 ubiquitin ligase anaphase-promoting complex. We also demonstrate that overexpression of FMRP can partially rescue hyperexcitability in TSC2-deficient iPSC-derived neurons. These data indicate that FMRP dysregulation represents an important pathological mechanism in the development of abnormal neuronal activity in TSC and illustrate a molecular convergence between these two neurogenetic disorders.
Collapse
Affiliation(s)
- Kellen D Winden
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Truc T Pham
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Nicole A Teaney
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Juan Ruiz
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ryan Chen
- Human Neuron Core, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Cidi Chen
- Human Neuron Core, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mustafa Sahin
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
4
|
Castano A, Silvestre M, Wells CI, Sanderson JL, Ferrer CA, Ong HW, Lang Y, Richardson W, Silvaroli JA, Bashore FM, Smith JL, Genereux IM, Dempster K, Drewry DH, Pabla NS, Bullock AN, Benke TA, Ultanir SK, Axtman AD. Discovery and characterization of a specific inhibitor of serine-threonine kinase cyclin-dependent kinase-like 5 (CDKL5) demonstrates role in hippocampal CA1 physiology. eLife 2023; 12:e88206. [PMID: 37490324 PMCID: PMC10406435 DOI: 10.7554/elife.88206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/24/2023] [Indexed: 07/26/2023] Open
Abstract
Pathological loss-of-function mutations in cyclin-dependent kinase-like 5 (CDKL5) cause CDKL5 deficiency disorder (CDD), a rare and severe neurodevelopmental disorder associated with severe and medically refractory early-life epilepsy, motor, cognitive, visual, and autonomic disturbances in the absence of any structural brain pathology. Analysis of genetic variants in CDD has indicated that CDKL5 kinase function is central to disease pathology. CDKL5 encodes a serine-threonine kinase with significant homology to GSK3β, which has also been linked to synaptic function. Further, Cdkl5 knock-out rodents have increased GSK3β activity and often increased long-term potentiation (LTP). Thus, development of a specific CDKL5 inhibitor must be careful to exclude cross-talk with GSK3β activity. We synthesized and characterized specific, high-affinity inhibitors of CDKL5 that do not have detectable activity for GSK3β. These compounds are very soluble in water but blood-brain barrier penetration is low. In rat hippocampal brain slices, acute inhibition of CDKL5 selectively reduces postsynaptic function of AMPA-type glutamate receptors in a dose-dependent manner. Acute inhibition of CDKL5 reduces hippocampal LTP. These studies provide new tools and insights into the role of CDKL5 as a newly appreciated key kinase necessary for synaptic plasticity. Comparisons to rodent knock-out studies suggest that compensatory changes have limited the understanding of the roles of CDKL5 in synaptic physiology, plasticity, and human neuropathology.
Collapse
Affiliation(s)
- Anna Castano
- Department of Pharmacology, University of Colorado School of MedicineAuroraUnited States
| | - Margaux Silvestre
- Kinases and Brain Development Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Carrow I Wells
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel HillChapel HillUnited States
| | - Jennifer L Sanderson
- Department of Pharmacology, University of Colorado School of MedicineAuroraUnited States
| | - Carla A Ferrer
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel HillChapel HillUnited States
| | - Han Wee Ong
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel HillChapel HillUnited States
| | - Yi Lang
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel HillChapel HillUnited States
| | - William Richardson
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Josie A Silvaroli
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State UniversityColumbusUnited States
| | - Frances M Bashore
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel HillChapel HillUnited States
| | - Jeffery L Smith
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel HillChapel HillUnited States
| | - Isabelle M Genereux
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel HillChapel HillUnited States
| | - Kelvin Dempster
- Kinases and Brain Development Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - David H Drewry
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel HillChapel HillUnited States
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel HillChapel HillUnited States
| | - Navlot S Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State UniversityColumbusUnited States
| | - Alex N Bullock
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Tim A Benke
- Departments of Pediatrics, Pharmacology, Neurology and Otolaryngology, University of Colorado School of MedicineAuroraUnited States
| | - Sila K Ultanir
- Kinases and Brain Development Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Alison D Axtman
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel HillChapel HillUnited States
| |
Collapse
|
5
|
Castano A, Silvestre M, Wells CI, Sanderson JL, Ferrer CA, Ong HW, Liang Y, Richardson W, Silvaroli JA, Bashore FM, Smith JL, Genereux IM, Dempster K, Drewry DH, Pabla NS, Bullock AN, Benke TA, Ultanir SK, Axtman AD. Discovery and characterization of a specific inhibitor of serine-threonine kinase cyclin dependent kinase-like 5 (CDKL5) demonstrates role in hippocampal CA1 physiology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.24.538049. [PMID: 37162893 PMCID: PMC10168277 DOI: 10.1101/2023.04.24.538049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Pathological loss-of-function mutations in cyclin-dependent kinase-like 5 ( CDKL5 ) cause CDKL5 deficiency disorder (CDD), a rare and severe neurodevelopmental disorder associated with severe and medically refractory early-life epilepsy, motor, cognitive, visual and autonomic disturbances in the absence of any structural brain pathology. Analysis of genetic variants in CDD have indicated that CDKL5 kinase function is central to disease pathology. CDKL5 encodes a serine-threonine kinase with significant homology to GSK3β, which has also been linked to synaptic function. Further, Cdkl5 knock-out rodents have increased GSK3β activity and often increased long-term potentiation (LTP). Thus, development of a specific CDKL5 inhibitor must be careful to exclude cross-talk with GSK3β activity. We synthesized and characterized specific, high-affinity inhibitors of CDKL5 that do not have detectable activity for GSK3β. These compounds are very soluble in water but blood-brain barrier penetration is low. In rat hippocampal brain slices, acute inhibition of CDKL5 selectively reduces post-synaptic function of AMPA-type glutamate receptors in a dose-dependent manner. Acute inhibition of CDKL5 reduces hippocampal LTP. These studies provide new tools and insights into the role of CDKL5 as a newly appreciated, key kinase necessary for synaptic plasticity. Comparisons to rodent knock-out studies suggest that compensatory changes have limited the understanding of the roles of CDKL5 in synaptic physiology, plasticity and human neuropathology.
Collapse
|
6
|
Remonde CG, Gonzales EL, Adil KJ, Jeon SJ, Shin CY. Augmented impulsive behavior in febrile seizure-induced mice. Toxicol Res 2023; 39:37-51. [PMID: 36726823 PMCID: PMC9839938 DOI: 10.1007/s43188-022-00145-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 04/04/2022] [Accepted: 07/01/2022] [Indexed: 02/04/2023] Open
Abstract
Febrile seizure (FS) is one of the most prevalent etiological events in childhood affecting 2-5% of children from 3 months to 5 years old. Debates on whether neurodevelopmental consequences rise in later life following a febrile seizure or not are still ongoing however there is limited evidence of its effect, especially in a laboratory setting. Moreover, the comparative study using both male and female animal models is sparse. To examine the effect of FS on the behavioral features of mice, both sexes of ICR mice were induced with hyperthermic seizures through exposure to an infrared heat lamp. The mice were divided into two groups, one receiving a single febrile seizure at postnatal day 11 (P11) and one receiving three FS at P11, P13, and P15. Starting at P30 the FS-induced mice were subjected to a series of behavioral tests. Mice with seizures showed no locomotor and motor coordination deficits, repetitive, and depressive-like behavior. However, the FS-induced mice showed impulsive-like behavior in both elevated plus maze and cliff avoidance tests, which is more prominent in male mice. A greater number of mice displayed impaired CAT in both males and females in the three-time FS-induced group compared to the single induction group. These results demonstrate that after induction of FS, male mice have a higher susceptibility to consequences of febrile seizure than female mice and recurrent febrile seizure has a higher chance of subsequent disorders associated with decreased anxiety and increased impulsivity. We confirmed the dysregulated expression of impulsivity-related genes such as 5-HT1A and tryptophan hydroxylase 2 from the prefrontal cortices of FS-induced mice implying that the 5-HT system would be one of the mechanisms underlying the increased impulsivity after FS. Taken together, these findings are useful in unveiling future discoveries about the effect of childhood febrile seizure and the mechanism behind it.
Collapse
Affiliation(s)
- Chilly Gay Remonde
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul, 05029 Republic of Korea
| | - Edson Luck Gonzales
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul, 05029 Republic of Korea
| | - Keremkleroo Jym Adil
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul, 05029 Republic of Korea
| | - Se Jin Jeon
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul, 05029 Republic of Korea
| | - Chan Young Shin
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul, 05029 Republic of Korea
| |
Collapse
|
7
|
Aishworiya R, Protic D, Hagerman R. Autism spectrum disorder in the fragile X premutation state: possible mechanisms and implications. J Neurol 2022; 269:4676-4683. [PMID: 35723724 DOI: 10.1007/s00415-022-11209-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 12/23/2022]
Abstract
There is increasing recognition of the heterogeneity of origin of cases of autism spectrum disorder (ASD) with multiple forms of ASD having been identified over the decades. Among these, a genetic etiology can be identified in 20-40% of cases when a full genetic work-up is completed. The Fragile X premutation state (characterized by the presence of 55-200 CGG repeats in the FMR1 gene) is a relatively newly identified disease state that has since been associated with several disorders including fragile X-associated tremor ataxia syndrome (FXTAS), fragile X-associated primary ovarian insufficiency (FXPOI) and most recently, fragile X-associated neurodevelopmental disorders (FXAND) which commonly includes anxiety and depression. In addition to these associated disorders, extant literature and clinical observations have suggested an association between the premutation state and ASD. In this paper, we review the literature pertinent to this and discuss possible molecular mechanisms that may explain this association. This includes lowered levels of the FMR1 Protein (FMRP), GABA deficits, mitochondrial dysfunction and secondary genetic abnormalities that is seen in premutation carriers as well as their increased vulnerability to environmental stressors. Understanding these mechanisms can facilitate development of targeted treatment for specific sub-groups of ASD and premutation disorders in future.
Collapse
Affiliation(s)
- Ramkumar Aishworiya
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA. .,Khoo Teck Puat-National University Children's Medical Institute, National University Health System, 5 Lower Kent Ridge Road, Singapore, 119074, Singapore. .,Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore, 117597, Singapore.
| | - Dragana Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Randi Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA.,Department of Pediatrics, University of California Davis School of Medicine, 4610 X St, Sacramento, CA, 95817, USA
| |
Collapse
|
8
|
Bigelow LJ, Fiset C, Jarvis JHM, Macleod S, Wöhr M, Benke TA, Bernard PB. Early-life seizures modify behavioral response to ultrasonic vocalization playback in adult rats. Epilepsy Behav 2022; 127:108494. [PMID: 34954511 DOI: 10.1016/j.yebeh.2021.108494] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022]
Abstract
Early-life seizures (ELS) are associated with autism spectrum disorder (ASD); however, due to a lack of effective treatments for ELS, it is not clear whether ELS plays a causal role, potentiates the ASD phenotype, or is the result of a common pathophysiology. Deficits in communications are a core feature of ASD. To isolate the impact of ELS on communication, we probed the behavioral consequences of a single episode of kainic acid-induced early-life seizures (KA-ELS) in male and female Sprague-Dawley (CD) rats. Deficits in auditory communication were observed in adult male rats as assessed by behavioral response to ultrasonic vocalization (USV) playback. Ultrasonic vocalizations are classified into two major categories - 50-kHz (positive) calls and 22-kHz (aversive) calls. Behavioral response was assessed via rat preference for different USV playback in a radial arm maze. Response to 22-kHz calls was not impacted by ELS while response to 50-kHz calls was impacted. All rats demonstrated positional preference for the arms adjacent to where 50-kHz calls were playing compared to background noise; however, male ELS rats demonstrated a greater positional preference for the arms adjacent to where 50-kHz calls were playing compared to male control rats. These studies demonstrate that responses to socially relevant auditory cues are chronically altered in adult male rats following a single episode of ELS. We speculate that these changes contribute to previously reported social deficits associated with ELS.
Collapse
Affiliation(s)
- Logan J Bigelow
- University of Prince Edward Island, Biomedical Sciences, Charlottetown, PE, Canada
| | - Catherine Fiset
- University of Prince Edward Island, Biomedical Sciences, Charlottetown, PE, Canada
| | - Jack H M Jarvis
- University of Prince Edward Island, Biomedical Sciences, Charlottetown, PE, Canada
| | - Sarah Macleod
- University of Prince Edward Island, Biomedical Sciences, Charlottetown, PE, Canada
| | - Markus Wöhr
- KU Leuven, Faculty of Psychology and Educational Sciences, Research Unit Brain and Cognition, Laboratory of Biological Psychology, Social and Affective Neuroscience Research Group, B-3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, B-3000 Leuven, Belgium; Behavioral Neuroscience, Experimental and Biological Psychology, Faculty of Psychology, Philipps University of Marburg, Gutenbergstr. 18, D-35032 Marburg, Germany; Center for Mind, Brain and Behavior, Philipps-University of Marburg, Hans-Meerwein-Str. 6, D-35032 Marburg, Germany
| | - Tim A Benke
- University of Colorado School of Medicine, Department of Pediatrics, Aurora, CO, United States
| | - Paul B Bernard
- University of Prince Edward Island, Biomedical Sciences, Charlottetown, PE, Canada.
| |
Collapse
|
9
|
Bleuzé L, Triaca V, Borreca A. FMRP-Driven Neuropathology in Autistic Spectrum Disorder and Alzheimer's disease: A Losing Game. Front Mol Biosci 2021; 8:699613. [PMID: 34760921 PMCID: PMC8573832 DOI: 10.3389/fmolb.2021.699613] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/24/2021] [Indexed: 12/28/2022] Open
Abstract
Fragile X mental retardation protein (FMRP) is an RNA binding protein (RBP) whose absence is essentially associated to Fragile X Syndrome (FXS). As an RNA Binding Protein (RBP), FMRP is able to bind and recognize different RNA structures and the control of specific mRNAs is important for neuronal synaptic plasticity. Perturbations of this pathway have been associated with the autistic spectrum. One of the FMRP partners is the APP mRNA, the main protagonist of Alzheimer’s disease (AD), thereby regulating its protein level and metabolism. Therefore FMRP is associated to two neurodevelopmental and age-related degenerative conditions, respectively FXS and AD. Although these pathologies are characterized by different features, they have been reported to share a number of common molecular and cellular players. The aim of this review is to describe the double-edged sword of FMRP in autism and AD, possibly allowing the elucidation of key shared underlying mechanisms and neuronal circuits. As an RBP, FMRP is able to regulate APP expression promoting the production of amyloid β fragments. Indeed, FXS patients show an increase of amyloid β load, typical of other neurological disorders, such as AD, Down syndrome, Parkinson’s Disease, etc. Beyond APP dysmetabolism, the two neurodegenerative conditions share molecular targets, brain circuits and related cognitive deficits. In this review, we will point out the potential common neuropathological pattern which needs to be addressed and we will hopefully contribute to clarifying the complex phenotype of these two neurorological disorders, in order to pave the way for a novel, common disease-modifying therapy.
Collapse
Affiliation(s)
- Louis Bleuzé
- University de Rennes 1, Rennes, France.,Humanitas Clinical and Research Center-IRCCS, Rozzano (Mi), Italy
| | - Viviana Triaca
- Institute of Biochemistry and Cell Biology, National Research Council (CNR-IBBC), International Campus A. Buzzati Traverso, Monterotondo, Italy
| | - Antonella Borreca
- Humanitas Clinical and Research Center-IRCCS, Rozzano (Mi), Italy.,Institute of Neuroscience-National Research Council (CNR-IN), Milan, Italy
| |
Collapse
|
10
|
A white paper on a neurodevelopmental framework for drug discovery in autism and other neurodevelopmental disorders. Eur Neuropsychopharmacol 2021; 48:49-88. [PMID: 33781629 DOI: 10.1016/j.euroneuro.2021.02.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/08/2021] [Accepted: 02/15/2021] [Indexed: 12/20/2022]
Abstract
In the last decade there has been a revolution in terms of genetic findings in neurodevelopmental disorders (NDDs), with many discoveries critical for understanding their aetiology and pathophysiology. Clinical trials in single-gene disorders such as fragile X syndrome highlight the challenges of investigating new drug targets in NDDs. Incorporating a developmental perspective into the process of drug development for NDDs could help to overcome some of the current difficulties in identifying and testing new treatments. This paper provides a summary of the proceedings of the 'New Frontiers Meeting' on neurodevelopmental disorders organised by the European College of Neuropsychopharmacology in conjunction with the Innovative Medicines Initiative-sponsored AIMS-2-TRIALS consortium. It brought together experts in developmental genetics, autism, NDDs, and clinical trials from academia and industry, regulators, patient and family associations, and other stakeholders. The meeting sought to provide a platform for focused communication on scientific insights, challenges, and methodologies that might be applicable to the development of CNS treatments from a neurodevelopmental perspective. Multidisciplinary translational consortia to develop basic and clinical research in parallel could be pivotal to advance knowledge in the field. Although implementation of clinical trials for NDDs in paediatric populations is widely acknowledged as essential, safety concerns should guide each aspect of their design. Industry and academia should join forces to improve knowledge of the biology of brain development, identify the optimal timing of interventions, and translate these findings into new drugs, allowing for the needs of users and families, with support from regulatory agencies.
Collapse
|
11
|
Berry-Kravis E, Filipink RA, Frye RE, Golla S, Morris SM, Andrews H, Choo TH, Kaufmann WE. Seizures in Fragile X Syndrome: Associations and Longitudinal Analysis of a Large Clinic-Based Cohort. Front Pediatr 2021; 9:736255. [PMID: 35036394 PMCID: PMC8756611 DOI: 10.3389/fped.2021.736255] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 10/07/2021] [Indexed: 11/15/2022] Open
Abstract
Fragile X syndrome (FXS), the most common inherited cause of intellectual disability, learning disability, and autism spectrum disorder, is associated with an increased prevalence of certain medical conditions including seizures. The goal of this study was to better understand seizures in individuals with FXS using the Fragile X Online Registry with Accessible Research Database, a multisite observational study initiated in 2012 involving FXS clinics in the Fragile X Clinic and Research Consortium. Seizure data were available for 1,607 participants, mostly male (77%) and white (74.5%). The overall prevalence of at least one seizure was 12%, with this rate being significantly higher in males than females (13.7 vs. 6.2%, p < 0.001). As compared to individuals with FXS without seizures, those with seizures were more likely to have autism spectrum disorder, current sleep apnea, later acquisition of expressive language, more severe intellectual disability, hyperactivity, irritability, and stereotyped movements. The mean age of seizure onset was 6.4 (SD 6.1) years of age with the great majority (>80%) having onset of seizures which was before 10. For those with epilepsy, about half (52%) had seizures for more than 3 years. This group was found to have greater cognitive and language impairment, but not behavioral disruptions, compared with those with seizures for <3 years. Antiepileptic drugs were more often used in males (60.6%) than females (34.8%), and females more often required more than one medication. The most commonly used anticonvulsants were oxcarbazepine, valproic acid, lamotrigine, and levetiracetam. The current study is the largest and first longitudinal study ever conducted to describe seizures in FXS. Overall, this study confirms previous reports of seizures in FXS and extends previous findings by further defining the cognitive and behavioral phenotype of those with epilepsy in FXS. Future studies should further investigate the natural history of seizures in FXS and the characteristics of seizures in FXS in adulthood.
Collapse
Affiliation(s)
- Elizabeth Berry-Kravis
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, United States.,Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States
| | - Robyn A Filipink
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Richard E Frye
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - Sailaja Golla
- Division of Neurodevelopmental Medicine, Department of Neurology, Thompson Autism Center, Children's Hospital of California, University of Irvine, Orange, CA, United States
| | - Stephanie M Morris
- Division of Pediatric and Developmental Neurology, Department of Neurology, Washington University in St. Louis, St. Louis, MO, United States
| | - Howard Andrews
- Department of Biostatistics, Mailman School of Public Health, Columbia University Medical Center, New York, NY, United States
| | - Tse-Hwei Choo
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY, United States
| | | | | |
Collapse
|
12
|
Increased expression of Fragile X mental retardation protein in malformative lesions of patients with focal cortical dysplasia. Neuroreport 2020; 31:1036-1041. [PMID: 32833881 DOI: 10.1097/wnr.0000000000001517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Focal cortical dysplasia (FCD) accounts for nearly half of all cases of medically refractory epilepsy in the pediatric and adult patient populations. This neurological disorder stems from localized malformations in cortical brain tissue due to impaired neuronal proliferation, differentiation, and migration patterns. Recent studies in animal models have highlighted the potential role of the Fragile X mental retardation protein (FMRP) levels in FCD. The purpose of this study was to investigate the status of FMRP activation in cortical brain tissues surgically resected from patients with FCD. In parallel, this study also investigated protein levels within the PI3K/AKT/mTOR and canonical Wnt signaling pathways. METHODS Pathologic tissue from malformative lesions of FCD patients with medically refractory epilepsy was compared to relatively normal control non-epileptic tissue from patients with intracranial neoplasms. A series of western blotting assays were performed to assess key proteins in the PI3K/AKT/mTOR, canonical Wnt signaling pathways, and FMRP. RESULTS There was suppression of S235/236-phosphorylated S6, GSK3α, and GSK3β protein levels in samples derived from FCD patients, compared to non-epileptic controls. FCD samples also had significantly greater levels of total and S499-phosphorylated FMRP. CONCLUSION These findings support our hypothesis that malformative lesions associated with FCD are characterized by high levels of FMRP activation along with dysregulation of both PI3K/AKT/mTOR and canonical Wnt signaling. These novel clinical findings extend previous work in animal models, further suggesting a potential unforeseen role of GSK3α and GSK3β in the pathophysiology of FCD and refractory epilepsy.
Collapse
|
13
|
O'Leary H, Vanderlinden L, Southard L, Castano A, Saba LM, Benke TA. Transcriptome analysis of rat dorsal hippocampal CA1 after an early life seizure induced by kainic acid. Epilepsy Res 2020; 161:106283. [PMID: 32062370 DOI: 10.1016/j.eplepsyres.2020.106283] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 01/17/2020] [Accepted: 01/29/2020] [Indexed: 12/13/2022]
Abstract
Seizures that occur during early development are associated with adverse neurodevelopmental outcomes. Causation and mechanisms are currently under investigation. Induction of an early life seizure by kainic acid (KA) in immature rats on post-natal day (P) 7 results in behavioral changes in the adult rat that reflect social and intellectual deficits without overt cellular damage. Our previous work also demonstrated increased expression of CA1 hippocampal long-term potentiation (LTP) and reduced desensitization of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type ionotropic glutamate receptors (AMPA-R) one week following a kainic acid induced seizure (KA-ELS). Here we used RNA sequencing (RNAseq) of mRNA from dorsal hippocampal CA1 to probe changes in mRNA levels one week following KA-ELS as a means to investigate the mechanisms for these functional changes. Ingenuity pathway analysis (IPA) confirmed our previous results by predicting an up-regulation of the synaptic LTP pathway. Differential gene expression results revealed significant differences in 7 gene isoforms. Additional assessments included AMPA-R splice variants and adenosine deaminase acting on RNA 2 (ADAR2) editing sites as a means to determine the mechanism for reduced AMPA-R desensitization. Splice variant analysis demonstrated that KA-ELS result in a small, but significant decrease in the "flop" isoform of Gria3, and editing site analysis revealed significant changes in the editing of a kainate receptor subunit, Grik2, and a serotonin receptor, Htr2c. While these specific changes may not account for altered AMPA-R desensitization, the differences indicate that KA-ELS alters gene expression in the hippocampal CA1 one week after the insult.
Collapse
Affiliation(s)
- Heather O'Leary
- Department of Pediatrics, University of Colorado, School of Medicine, 80045, United States.
| | - Lauren Vanderlinden
- Department of Biostatistics and Informatics, Colorado School of Public Health, 80045, United States.
| | - Lara Southard
- Department of Psychology, Colorado State University, Fort Collins, 80523, United States.
| | - Anna Castano
- Department of Pediatrics, University of Colorado, School of Medicine, 80045, United States.
| | - Laura M Saba
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, 80045, United States.
| | - Tim A Benke
- Department of Pediatrics, University of Colorado, School of Medicine, 80045, United States; Department of Neurology, University of Colorado, School of Medicine, 80045, United States; Department of Pharmacology, University of Colorado, School of Medicine, 80045, United States; Department of Otolaryngology, University of Colorado, School of Medicine, 80045, United States; Neuroscience Graduate Program, University of Colorado, School of Medicine, 80045, United States.
| |
Collapse
|
14
|
Kim K, Hessl D, Randol JL, Espinal GM, Schneider A, Protic D, Aydin EY, Hagerman RJ, Hagerman PJ. Association between IQ and FMR1 protein (FMRP) across the spectrum of CGG repeat expansions. PLoS One 2019; 14:e0226811. [PMID: 31891607 PMCID: PMC6938341 DOI: 10.1371/journal.pone.0226811] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/05/2019] [Indexed: 12/20/2022] Open
Abstract
Fragile X syndrome, the leading heritable form of intellectual disability, is caused by hypermethylation and transcriptional silencing of large (CGG) repeat expansions (> 200 repeats) in the 5′ untranslated region of the fragile X mental retardation 1 (FMR1) gene. As a consequence of FMR1 gene silencing, there is little or no production of FMR1 protein (FMRP), an important element in normal synaptic function. Although the absence of FMRP has long been known to be responsible for the cognitive impairment in fragile X syndrome, the relationship between FMRP level and cognitive ability (IQ) is only imprecisely understood. To address this issue, a high-throughput, fluorescence resonance energy transfer (FRET) assay has been used to quantify FMRP levels in dermal fibroblasts, and the relationship between FMRP and IQ measures was assessed by statistical analysis in a cohort of 184 individuals with CGG-repeat lengths spanning normal (< 45 CGGs) to full mutation (> 200 CGGs) repeat ranges in fibroblasts. The principal findings of the current study are twofold: i) For those with normal CGG repeats, IQ is no longer sensitive to further increases in FMRP above an FMRP threshold of ~70% of the mean FMRP level; below this threshold, IQ decreases steeply with further decreases in FMRP; and ii) For the current cohort, a mean IQ of 85 (lower bound for the normal IQ range) is attained for FMRP levels that are only ~35% of the mean FMRP level among normal CGG-repeat controls. The current results should help guide expectations for efforts to induce FMR1 gene activity and for the levels of cognitive function expected for a given range of FMRP levels.
Collapse
Affiliation(s)
- Kyoungmi Kim
- UC Davis MIND Institute, UC Davis Health, Sacramento, California, United States of America
- Department of Public Health Sciences, University of California, Davis, School of Medicine, Davis, California, United States of America
| | - David Hessl
- UC Davis MIND Institute, UC Davis Health, Sacramento, California, United States of America
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, School of Medicine, Sacramento, California, United States of America
| | - Jamie L. Randol
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, California, United States of America
| | - Glenda M. Espinal
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, California, United States of America
| | - Andrea Schneider
- UC Davis MIND Institute, UC Davis Health, Sacramento, California, United States of America
- Department of Pediatrics, University of California, Davis, School of Medicine, Sacramento, California, United States of America
| | - Dragana Protic
- UC Davis MIND Institute, UC Davis Health, Sacramento, California, United States of America
| | - Elber Yuksel Aydin
- UC Davis MIND Institute, UC Davis Health, Sacramento, California, United States of America
| | - Randi J. Hagerman
- UC Davis MIND Institute, UC Davis Health, Sacramento, California, United States of America
- Department of Pediatrics, University of California, Davis, School of Medicine, Sacramento, California, United States of America
| | - Paul J. Hagerman
- UC Davis MIND Institute, UC Davis Health, Sacramento, California, United States of America
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
15
|
EEG Abnormalities as a Neurophysiological Biomarker of Severity in Autism Spectrum Disorder: A Pilot Cohort Study. J Autism Dev Disord 2019; 49:2337-2347. [PMID: 30726535 DOI: 10.1007/s10803-019-03908-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
To date, the phenotypic significance of EEG abnormalities in patients with ASD is unclear. In a population affected by ASD we aimed to evaluate: the phenotypic characteristics; the prevalence of EEG abnormalities; the potential correlations between EEG abnormalities and behavioral and cognitive variables. Sixty-nine patients with ASD underwent cognitive or developmental testing, language assessment, and adaptive behavior skills evaluation as well as sleep/wake EEG recording. EEG abnormalities were found in 39.13% of patients. EEG abnormalities correlated with autism severity, hyperactivity, anger outbursts, aggression, negative or destructive behavior, motor stereotypies, intellectual disability, language impairment and self-harm. Our findings confirmed that EEG abnormalities are present in the ASD population and correlate with several associated phenotypic features.
Collapse
|
16
|
Protein Translation in the Nucleus Accumbens Is Dysregulated during Cocaine Withdrawal and Required for Expression of Incubation of Cocaine Craving. J Neurosci 2018; 38:2683-2697. [PMID: 29431650 DOI: 10.1523/jneurosci.2412-17.2018] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 01/08/2018] [Accepted: 01/31/2018] [Indexed: 01/11/2023] Open
Abstract
Exposure to drug-associated cues can induce drug craving and relapse in abstinent addicts. Cue-induced craving that progressively intensifies ("incubates") during withdrawal from cocaine has been observed in both rats and humans. Building on recent evidence that aberrant protein translation underlies incubation-related adaptations in the NAc, we used male rats to test the hypothesis that translation is dysregulated during cocaine withdrawal and/or when rats express incubated cocaine craving. We found that intra-NAc infusion of anisomycin, a general protein translation inhibitor, or rapamycin, an inhibitor of mammalian target of rapamycin, reduced the expression of incubated cocaine craving, consistent with previous results showing that inhibition of translation in slices normalized the adaptations that maintain incubation. We then examined signaling pathways involved in protein translation using NAc synaptoneurosomes prepared after >47 d of withdrawal from cocaine or saline self-administration, or after withdrawal plus a cue-induced seeking test. The most robust changes were observed following seeking tests. Most notably, we found that eukaryotic elongation factor 2 (eEF2) and eukaryotic initiation factor 2α (eIF2α) are dephosphorylated when cocaine rats undergo a cue-induced seeking test; both effects are consistent with increased translation during the test. Blocking eIF2α dephosphorylation and thereby restoring its inhibitory influence on translation, via intra-NAc injection of Sal003 just before the test, substantially reduced cocaine seeking. These results are consistent with dysregulation of protein translation in the NAc during cocaine withdrawal, enabling cocaine cues to elicit an aberrant increase in translation that is required for the expression of incubated cocaine craving.SIGNIFICANCE STATEMENT Cue-induced cocaine craving progressively intensifies (incubates) during withdrawal in both humans and rats. This may contribute to persistent vulnerability to relapse. We previously demonstrated a role for protein translation in synaptic adaptations in the NAc closely linked to incubation. Here, we tested the hypothesis that translation is dysregulated during cocaine withdrawal, and this contributes to incubated craving. Analysis of signaling pathways regulating translation suggested that translation is enhanced when "incubated" rats undergo a cue-induced seeking test. Furthermore, intra-NAc infusions of drugs that inhibit protein translation through different mechanisms reduced expression of incubated cue-induced cocaine seeking. These results demonstrate that the expression of incubation depends on an acute increase in translation that may result from dysregulation of several pathways.
Collapse
|
17
|
|
18
|
Wheeler A, Raspa M, Hagerman R, Mailick M, Riley C. Implications of the FMR1 Premutation for Children, Adolescents, Adults, and Their Families. Pediatrics 2017; 139:S172-S182. [PMID: 28814538 PMCID: PMC5621635 DOI: 10.1542/peds.2016-1159d] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/24/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Given the nature of FMR1 gene expansions, most biological mothers, and often multiple other family members of children with fragile X syndrome (FXS), will have a premutation, which may increase individual and family vulnerabilities. This article summarizes important gaps in knowledge and notes potential implications for pediatric providers with regard to developmental and medical risks for children and adolescents with an FMR1 premutation, including possible implications into adulthood. METHODS A structured electronic literature search was conducted on FMR1 pre- and full mutations, yielding a total of 306 articles examined. Of these, 116 focused primarily on the premutation and are included in this review. RESULTS Based on the literature review, 5 topic areas are discussed: genetics and epidemiology; phenotypic characteristics of individuals with the premutation; implications for carrier parents of children with FXS; implications for the extended family; and implications for pediatricians. CONCLUSIONS Although the premutation phenotype is typically less severe in clinical presentation than in FXS, premutation carriers are much more common and are therefore more likely to be seen in a typical pediatric practice. In addition, there is a wide range of medical, cognitive/developmental, and psychiatric associated features that individuals with a premutation are at increased risk for having, which underscores the importance of awareness on the part of pediatricians in identifying and monitoring premutation carriers and recognizing the impact this identification may have on family members.
Collapse
Affiliation(s)
- Anne Wheeler
- RTI International, Research Triangle Park, North Carolina;
| | - Melissa Raspa
- RTI International, Research Triangle Park, North Carolina
| | - Randi Hagerman
- MIND Institute, University of California at Davis, Sacramento, California
| | - Marsha Mailick
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin; and
| | - Catharine Riley
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
19
|
The Impact of Electrographic Seizures on Developing Hippocampal Dendrites Is Calcineurin Dependent. eNeuro 2017; 4:eN-NWR-0014-17. [PMID: 28462391 PMCID: PMC5409981 DOI: 10.1523/eneuro.0014-17.2017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/01/2017] [Accepted: 03/03/2017] [Indexed: 11/21/2022] Open
Abstract
Neurobehavioral abnormalities are commonly associated with intractable childhood epilepsy. Studies from numerous labs have demonstrated cognitive and socialization deficits in rats and mice that have experienced early-life seizures. However, the cellular and molecular mechanisms underlying these effects are unknown. Previously, experiments have shown that recurrent seizures in infancy suppress the growth of hippocampal dendrites at the same time they impair learning and memory. Experiments in slice cultures have also demonstrated dendrite growth suppression. Here, we crossed calcineurin B1 (CaNB1) floxed and Thy1GFP-M mice to produce mice that were homozygous for the both the floxed CaNB1 and the Thy1GFP-M transgene. Littermates that were homozygous for wild-type CaNB1 and Thy1GFP-M served as controls. Hippocampal slice cultures from these mice were transfected with an AAV/hSyn-mCherry-Cre virus to eliminate CaNB1 from neurons. Immunohistochemical results showed that CaNB1 was eliminated from at least 90% of the transfected CA1 pyramidal cells. Moreover, the CaN-dependent nuclear translocation of the CREB transcription coactivator, CREB-regulated transcriptional coactivator 1 (CRTC1), was blocked in transfected neurons. Cell attach patch recordings combined with live multiphoton imaging demonstrated that the loss of CaNB1 did not prevent neurons from fully participating in electrographic seizure activity. Finally, dendrite reconstruction showed that the elimination of CaNB1 prevented seizure-induced decreases in both dendrite length and branch number. Results suggest that CaN plays a key role in seizure-induced dendrite growth suppression and may contribute to the neurobehavioral comorbidities of childhood epilepsy.
Collapse
|
20
|
Smith G, Ahmed N, Arbuckle E, Lugo JN. Early-life status epilepticus induces long-term deficits in anxiety and spatial learning in mice. INTERNATIONAL JOURNAL OF EPILEPSY 2017; 4:36-45. [PMID: 31890565 PMCID: PMC6936764 DOI: 10.1016/j.ijep.2016.12.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND One of the most devastating aspects of developmental epilepsy is the long-term impact on behavior. Children with epilepsy show a high co-morbidity with anxiety disorders and autism. METHODS To examine whether early-life status epilepticus results in altered anxiety, repetitive behavior, social behavior, and learning and memory, we induced status epilepticus in male C57BL/6 mice on postnatal day (PD) 10. The mice received intraperitoneal injections of either kainic acid (2mg/kg) or 0.9% normal saline. We also included a nontreated control group. Kainic acid induced status epilepticus for approximately 1.5 hrs. At PD60, the adult mice were then tested in a battery of behavioral tasks, including open field activity, elevated-plus maze, light-dark test, marble burying, social chamber, social partition, conditioned fear, novel object recognition, and Morris water maze. RESULTS The early-life seizure group showed consistent increases in anxiety in the open field test (p < 0.05), elevated plus maze (p < 0.05), and light-dark task (p < 0.01). The seizure group showed significant (p < 0.01) impairment in the Morris water maze. There were no differences observed in marble burying, social partition, social chamber, novel object recognition, or delay fear conditioning tasks. CONCLUSIONS These results demonstrate that a single insult of status epilepticus during the neonatal period is sufficient to cause specific, long-term impairments in anxiety and spatial learning.
Collapse
Affiliation(s)
- Gregory Smith
- Institute of Biomedical Sciences, Baylor University, Waco, TX 76798, USA
| | - Nowrin Ahmed
- Department of Psychology and Neuroscience, Baylor University, Waco, TX 76798, USA
| | - Erin Arbuckle
- Institute of Biomedical Sciences, Baylor University, Waco, TX 76798, USA
| | - Joaquin N. Lugo
- Institute of Biomedical Sciences, Baylor University, Waco, TX 76798, USA
- Department of Psychology and Neuroscience, Baylor University, Waco, TX 76798, USA
| |
Collapse
|
21
|
Superimposing Status Epilepticus on Neuron Subset-Specific PTEN Haploinsufficient and Wild Type Mice Results in Long-term Changes in Behavior. Sci Rep 2016; 6:36559. [PMID: 27819284 PMCID: PMC5098193 DOI: 10.1038/srep36559] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 10/18/2016] [Indexed: 01/09/2023] Open
Abstract
We evaluated the effects of superimposing seizures on a genetic mutation with known involvement in both Autism Spectrum Disorder and in epilepsy. Neuron-subset specific (NS)-Pten heterozygous (HT) and wildtype (WT) adult mice received either intraperitoneal injections of kainic acid (20 mg/kg) to induce status epilepticus or the vehicle (saline). Animals then received a battery of behavioral tasks in order to evaluate activity levels, anxiety, repetitive-stereotyped behavior, social behavior, learning and memory. In the open field task, we found that HT mice after seizures showed a significant increase in total activity and total distance in the surround region of the open field. In the elevated plus maze task, we found that HT mice after seizures displayed increased total distance and velocity as compared to HT mice that did not undergo seizures and WT controls. In the social chamber test, we found the HT mice after seizures displayed an impairment in social behavior. These findings demonstrate that superimposing seizures on a genetic mutation can result in long-term alterations in activity and social behavior in mice.
Collapse
|
22
|
Han T, Qin Y, Mou C, Wang M, Jiang M, Liu B. Seizure induced synaptic plasticity alteration in hippocampus is mediated by IL-1β receptor through PI3K/Akt pathway. Am J Transl Res 2016; 8:4499-4509. [PMID: 27830035 PMCID: PMC5095344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 10/02/2016] [Indexed: 06/06/2023]
Abstract
Seizures, which result from synchronized aberrant firing of neuronal populations, can cause long-term sequelae, such as epilepsy, cognitive and behavioral issues, in which the synaptic plasticity alteration may play an important role. Long-term potentiation (LTP) is a persistent increase in synaptic strength and is essential for learning and memory. In the present study, we first examined the alteration of cognitive impairments and synaptic plasticity in mice with seizures, then explored the underlying mechanism involving pro-inflammatory factors and PI3K/Akt pathway. The results demonstrated that: (1) PTZ-induced seizure impairs learning and memory in mice, indicated by Morris water maze test; (2) PTZ-induced seizure decreased LTP; (3) the mRNA expression of IL-1β, IL-6 and TNF-α in the hippocampus were increased in mice with seizures; (4) LTP was increased by IL-1β receptor antagonist anakinra, but not inhibitors of IL-6 or TNF-α receptor; (5) Antagonist of IL-1β receptor rescues deficits in learning and memory of mice with seizures through PI3K/Akt pathway. It is concluded that the IL-1β induced by PTZ-induced seizures may impair the synaptic plasticity alteration in hippocampus as well as learning and memory ability by PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Tao Han
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University 9677 Jingshi Road, Jinan, Shandong 250021, China
| | - Yanyu Qin
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University 9677 Jingshi Road, Jinan, Shandong 250021, China
| | - Chenzhi Mou
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University 9677 Jingshi Road, Jinan, Shandong 250021, China
| | - Min Wang
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University 9677 Jingshi Road, Jinan, Shandong 250021, China
| | - Meng Jiang
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University 9677 Jingshi Road, Jinan, Shandong 250021, China
| | - Bin Liu
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University 9677 Jingshi Road, Jinan, Shandong 250021, China
| |
Collapse
|
23
|
Reynolds CD, Smith G, Jefferson T, Lugo JN. The effect of early life status epilepticus on ultrasonic vocalizations in mice. Epilepsia 2016; 57:1377-85. [PMID: 27378279 DOI: 10.1111/epi.13450] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2016] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Infant crying is a series of innate vocal patterns intended to elicit the attention of adult caregivers for fulfillment of specific needs such as pain, hunger, or hypostimulation. It is one of the earliest forms of observable communication. In neonatal rodents, this behavior has recently been investigated as a potential early behavioral marker of neural deficits in neurodevelopmental disorders. However, few studies have examined the effects of seizures on vocalization behavior during the neonatal period. The purpose of this study is to investigate the effect of a single kainate-induced early life seizure on vocalization behavior in mice. This study also investigates the subsequent effect of seizures on two pathways critical for early neural development and epileptogenesis: the phosphoinositide 3-kinase|serine/threonine kinase|mammalian target of rapamycin (PI3K-Akt-mTOR) and canonical (Wingless-Int Wnt) intracellular signaling pathways. METHODS On postnatal day 10, male and female 129SvEvTac mice received a single intraperitoneal injection of kainic acid (2.5 mg/kg) or vehicle injection. The kainate administration resulted in 1-2 h of status epilepticus. On postnatal days 11 and 12, the quantity and duration of isolation-induced ultrasonic vocalizations were recorded. Western blotting analyses were performed using male and female pups on postnatal day 12. RESULTS There was significant, male-specific suppression in the quantity and total duration of 50-kHz calls on postnatal day 12 following seizures. The hippocampi of male mice on this postnatal day also revealed male-specific changes in the PI3K-Akt-mTOR intracellular signaling pathway, as well as changes in phosphorylated fragile × mental retardation protein. SIGNIFICANCE These findings demonstrate that early life seizures can disrupt communication behavior in neonatal mice.
Collapse
Affiliation(s)
- Conner D Reynolds
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, U.S.A
| | - Gregory Smith
- Institute of Biomedical Sciences, Baylor University, Waco, Texas, U.S.A
| | - Taylor Jefferson
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, U.S.A
| | - Joaquin N Lugo
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, U.S.A.,Institute of Biomedical Sciences, Baylor University, Waco, Texas, U.S.A
| |
Collapse
|
24
|
O'Leary H, Bernard PB, Castano AM, Benke TA. Enhanced long term potentiation and decreased AMPA receptor desensitization in the acute period following a single kainate induced early life seizure. Neurobiol Dis 2015; 87:134-44. [PMID: 26706598 DOI: 10.1016/j.nbd.2015.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 11/17/2015] [Accepted: 12/14/2015] [Indexed: 02/04/2023] Open
Abstract
Neonatal seizures are associated with long term disabilities including epilepsy and cognitive deficits. Using a neonatal seizure rat model that does not develop epilepsy, but develops a phenotype consistent with other models of intellectual disability (ID) and autism spectrum disorders (ASD), we sought to isolate the acute effects of a single episode of early life seizure on hippocampal CA1 synaptic development and plasticity. We have previously shown chronic changes in glutamatergic synapses, loss of long term potentiation (LTP) and enhanced long term depression (LTD), in the adult male rat ~50days following kainic acid (KA) induced early life seizure (KA-ELS) in post-natal (P) 7day old male Sprague-Dawley rats. In the present work, we examined the electrophysiological properties and expression levels of glutamate receptors in the acute period, 2 and 7days, post KA-ELS. Our results show for the first time enhanced LTP 7days after KA-ELS, but no change 2days post KA-ELS. Additionally, we report that ionotropic α-amino-3-hydroxy-5-methyl-isoxazole-propionic acid type glutamate receptor (AMPAR) desensitization is decreased in the same time frame, with no changes in AMPAR expression, phosphorylation, or membrane insertion. Inappropriate enhancement of the synaptic connections in the acute period after the seizure could alter the normal patterning of synaptic development in the hippocampus during this critical period and contribute to learning deficits. Thus, this study demonstrates a novel mechanism by which KA-ELS alters early network properties that potentially lead to adverse outcomes.
Collapse
Affiliation(s)
- Heather O'Leary
- Department of Pediatrics, University of Colorado, School of Medicine, 80045, USA
| | - Paul B Bernard
- Department of Pediatrics, University of Colorado, School of Medicine, 80045, USA
| | - Anna M Castano
- Department of Pediatrics, University of Colorado, School of Medicine, 80045, USA
| | - Tim A Benke
- Department of Pediatrics, University of Colorado, School of Medicine, 80045, USA; Department of Neurology, University of Colorado, School of Medicine, 80045, USA; Department of Pharmacology, University of Colorado, School of Medicine, 80045, USA; Department of Otolaryngology, University of Colorado, School of Medicine, 80045, USA; Neuroscience Graduate Program, University of Colorado, School of Medicine, 80045, USA.
| |
Collapse
|
25
|
Biever A, Valjent E, Puighermanal E. Ribosomal Protein S6 Phosphorylation in the Nervous System: From Regulation to Function. Front Mol Neurosci 2015; 8:75. [PMID: 26733799 PMCID: PMC4679984 DOI: 10.3389/fnmol.2015.00075] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/23/2015] [Indexed: 01/31/2023] Open
Abstract
Since the discovery of the phosphorylation of the 40S ribosomal protein S6 (rpS6) about four decades ago, much effort has been made to uncover the molecular mechanisms underlying the regulation of this post-translational modification. In the field of neuroscience, rpS6 phosphorylation is commonly used as a readout of the mammalian target of rapamycin complex 1 signaling activation or as a marker for neuronal activity. Nevertheless, its biological role in neurons still remains puzzling. Here we review the pharmacological and physiological stimuli regulating this modification in the nervous system as well as the pathways that transduce these signals into rpS6 phosphorylation. Altered rpS6 phosphorylation observed in various genetic and pathophysiological mouse models is also discussed. Finally, we examine the current state of knowledge on the physiological role of this post-translational modification and highlight the questions that remain to be addressed.
Collapse
Affiliation(s)
- Anne Biever
- Centre National de la Recherche Scientifique, UMR5203, Institut de Génomique FonctionnelleMontpellier, France; Institut National de la Santé et de la Recherche Médicale, U1191Montpellier, France; Université de Montpellier, UMR-5203Montpellier, France
| | - Emmanuel Valjent
- Centre National de la Recherche Scientifique, UMR5203, Institut de Génomique FonctionnelleMontpellier, France; Institut National de la Santé et de la Recherche Médicale, U1191Montpellier, France; Université de Montpellier, UMR-5203Montpellier, France
| | - Emma Puighermanal
- Centre National de la Recherche Scientifique, UMR5203, Institut de Génomique FonctionnelleMontpellier, France; Institut National de la Santé et de la Recherche Médicale, U1191Montpellier, France; Université de Montpellier, UMR-5203Montpellier, France
| |
Collapse
|
26
|
Reynolds CD, Smith GD, Jefferson TS, Lugo JN. Comparison of Equivalence between Two Commercially Available S499-Phosphorylated FMRP Antibodies in Mice. PLoS One 2015; 10:e0143134. [PMID: 26580204 PMCID: PMC4651511 DOI: 10.1371/journal.pone.0143134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 10/31/2015] [Indexed: 11/18/2022] Open
Abstract
Fragile X syndrome (FXS) develops from excessive trinucleotide CGG repeats in the 5’-untranslated region at Xq27.3 of the Fmr-1 gene, which functionally silences its expression and prevents transcription of its protein. This disorder is the most prominent form of heritable intellectual deficiency, affecting roughly 1 in 5,000 males and 1 in 10,000 females globally. Antibody specificity and selectivity are essential for investigating changes in intracellular protein signaling and phosphorylation status of the Fragile X Mental Retardation Protein (FMRP). Currently, both PhosphoSolutions® and abcam® produce commercially available S499-phosphorylated FMRP specific antibodies. The antibody from PhosphoSolutions® has been validated in previous studies; however, the antibody from abcam® antibody has yet to receive similar validation. This study aims to determine whether these two antibodies are true equivalents through western blot analysis of both NS-Pten knockout (KO) and Fmr-1 KO mice strains. We prepared hippocampal synaptosomal preparations and probed the samples using total FMRP, abcam® phosphorylated FMRP, and PhosphoSolutions® phosphorylated FMRP antibodies. We found that there was a significant increase in phosphorylated FMRP levels using the abcam® and PhosphoSolutions® antibodies in the NS-Pten KO mice compared to wildtype mice. However, there was much more variability using the abcam® antibody. Furthermore, there was a band present in the Fmr-1 KO for the phosphorylated FMRP site using the abcam® antibody for western blotting but not for the PhosphoSolutions® antibody. Our findings strongly suggest that the antibody from abcam® is neither specific nor selective for its advertised targeted substrate, S499-phosphorylated FMRP.
Collapse
Affiliation(s)
- Conner D. Reynolds
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, United States of America
| | - Gregory D. Smith
- Institute of Biomedical Sciences, Baylor University, Waco, Texas, United States of America
| | - Taylor S. Jefferson
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, United States of America
| | - Joaquin N. Lugo
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, United States of America
- Institute of Biomedical Sciences, Baylor University, Waco, Texas, United States of America
- * E-mail:
| |
Collapse
|
27
|
Opportunities for improving animal welfare in rodent models of epilepsy and seizures. J Neurosci Methods 2015; 260:2-25. [PMID: 26376175 DOI: 10.1016/j.jneumeth.2015.09.007] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 09/01/2015] [Accepted: 09/08/2015] [Indexed: 01/31/2023]
Abstract
Animal models of epilepsy and seizures, mostly involving mice and rats, are used to understand the pathophysiology of the different forms of epilepsy and their comorbidities, to identify biomarkers, and to discover new antiepileptic drugs and treatments for comorbidities. Such models represent an important area for application of the 3Rs (replacement, reduction and refinement of animal use). This report provides background information and recommendations aimed at minimising pain, suffering and distress in rodent models of epilepsy and seizures in order to improve animal welfare and optimise the quality of studies in this area. The report includes practical guidance on principles of choosing a model, induction procedures, in vivo recordings, perioperative care, welfare assessment, humane endpoints, social housing, environmental enrichment, reporting of studies and data sharing. In addition, some model-specific welfare considerations are discussed, and data gaps and areas for further research are identified. The guidance is based upon a systematic review of the scientific literature, survey of the international epilepsy research community, consultation with veterinarians and animal care and welfare officers, and the expert opinion and practical experience of the members of a Working Group convened by the United Kingdom's National Centre for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs).
Collapse
|
28
|
Winarni TI, Schneider A, Ghaziuddin N, Seritan A, Hagerman RJ. Psychosis and catatonia in fragile X: Case report and literature review. Intractable Rare Dis Res 2015; 4:139-46. [PMID: 26361565 PMCID: PMC4561243 DOI: 10.5582/irdr.2015.01028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 07/31/2015] [Accepted: 08/12/2015] [Indexed: 12/13/2022] Open
Abstract
Fragile X mental retardation 1 (FMR1) premutation associated phenotypes have been explored extensively since the molecular mechanism emerged involving elevated FMR1 messenger ribonucleic acid (mRNA) levels. Lowered fragile X mental retardation protein (FMRP) can also occur which may have an additive effect to the high levels of mRNA leading to neurodevelopmental problems and psychopathology. This paper was aimed to review psychosis and catatonia in premutation carriers, express the role of elevated FMR1 mRNA and lowered FMRP in the phenotype of carriers and present a case of psychosis and catatonia in a carrier. This case also demonstrates additional genetic and environmental factors which may also affect the phenotype. We review the literature and report an exemplary case of a 25 year old male premutation carrier with elevated FMR1 mRNA, low FMRP, a cytochrome P450 family 2 subfamily D polypeptide 6 (CYP2D6)*2xN mutation and a perinatal insult. This patient developed an autism spectrum disorder, psychosis, catatonia with subsequent cognitive decline after electro-convulsive therapy (ECT) for his catatonia. He had a premutation of 72 CGG repeat in FMR1, FMR1 mRNA level that was over 2.4 times normal and FMRP level at 18% of normal, and additionally, a CYP2D6 allelic variant which leads to ultrarapid metabolism (UM) of medication. There is an overlapping pathophysiological mechanism of catatonia and fragile X-associated premutation phenotypes including autism and psychosis. This case demonstrates the shared phenotype and the overlap of the pathophysiological mechanisms that can influence the intervention. Multiple genetic and environmental hits can lead to more significant involvement in premutation carriers.
Collapse
Affiliation(s)
- Tri Indah Winarni
- MIND Institute, University of California Davis, Medical Center, Sacramento, USA
- Center for Biomedical Research (CEBIOR), Faculty of Medicine Diponegoro University, Semarang, Indonesia
| | - Andrea Schneider
- MIND Institute, University of California Davis, Medical Center, Sacramento, USA
- Department of Pediatrics, University of California Davis, Medical Center, Sacramento, USA
| | - Neera Ghaziuddin
- University of Michigan Hospitals and Health Center, Ann Arbor, USA
| | - Andreea Seritan
- Department of Psychiatry and Behavioral Sciences, University of California Davis, Medical Center, Sacramento, USA
| | - Randi J Hagerman
- MIND Institute, University of California Davis, Medical Center, Sacramento, USA
- Department of Pediatrics, University of California Davis, Medical Center, Sacramento, USA
- Address correspondence to: Dr. Randi J. Hagerman, MIND Institute, UC Davis Health System, 2825 50th Street, Sacramento, CA 95817, USA. E-mail:
| |
Collapse
|
29
|
Modulation of the genome and epigenome of individuals susceptible to autism by environmental risk factors. Int J Mol Sci 2015; 16:8699-718. [PMID: 25903146 PMCID: PMC4425104 DOI: 10.3390/ijms16048699] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 04/03/2015] [Accepted: 04/08/2015] [Indexed: 12/12/2022] Open
Abstract
Diverse environmental factors have been implicated with the development of autism spectrum disorders (ASD). Genetic factors also underlie the differential vulnerability to environmental risk factors of susceptible individuals. Currently the way in which environmental risk factors interact with genetic factors to increase the incidence of ASD is not well understood. A greater understanding of the metabolic, cellular, and biochemical events involved in gene x environment interactions in ASD would have important implications for the prevention and possible treatment of the disorder. In this review we discuss various established and more alternative processes through which environmental factors implicated in ASD can modulate the genome and epigenome of genetically-susceptible individuals.
Collapse
|
30
|
Hagerman PJ, Hagerman RJ. Fragile X-associated tremor/ataxia syndrome. Ann N Y Acad Sci 2015; 1338:58-70. [PMID: 25622649 PMCID: PMC4363162 DOI: 10.1111/nyas.12693] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 12/04/2014] [Accepted: 12/18/2014] [Indexed: 12/20/2022]
Abstract
Fragile X-associated tremor/ataxia syndrome (FXTAS) is a late-onset neurodegenerative disorder that affects some but not all carriers of small, noncoding CGG-repeat expansions (55-200 repeats; premutation) within the fragile X gene (FMR1). Principal features of FXTAS include intention tremor, cerebellar ataxia, Parkinsonism, memory and executive function deficits, autonomic dysfunction, brain atrophy with white matter disease, and cognitive decline. Although FXTAS was originally considered to be confined to the premutation range, rare individuals with a gray zone (45-54 repeats) or an unmethylated full mutation (>200 repeats) allele have now been described, the constant feature of the disorder remaining the requirement for FMR1 expression, in contradistinction to the gene silencing mechanism of fragile X syndrome. Although transcriptional activity is required for FXTAS pathogenesis, the specific trigger(s) for FXTAS pathogenesis remains elusive, highlighting the need for more research in this area. This need is underscored by recent neuroimaging findings of changes in the central nervous system that consistently appear well before the onset of clinical symptoms, thus creating an opportunity to delay or prevent the appearance of FXTAS.
Collapse
Affiliation(s)
- Paul J Hagerman
- Department of Biochemistry and Molecular Medicine, University of California , Davis , School of Medicine, Davis, California; The MIND Institute, University of California , Davis , Health System, Sacramento, California
| | | |
Collapse
|
31
|
Bernard PB, Castano AM, Beitzel CS, Carlson VB, Benke TA. Behavioral changes following a single episode of early-life seizures support the latent development of an autistic phenotype. Epilepsy Behav 2015; 44:78-85. [PMID: 25659043 PMCID: PMC4405461 DOI: 10.1016/j.yebeh.2015.01.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 01/06/2015] [Accepted: 01/07/2015] [Indexed: 01/23/2023]
Abstract
We probed the developmental and behavioral consequences of a single episode of kainic acid-induced early-life seizures (KA-ELS) in the rat on postnatal day 7. Correlates of developmental trajectory were not altered, demonstrating that long-term consequences following KA-ELS are not initiated by secondary causes, such as malnourishment or alterations in maternal care. We report reduced marble burying in adult rats, suggestive of restricted interests, a trait common to experimental and clinical autism. We did not detect increased repetitive grooming during habituated cage behavior. However, we did detect reduced grooming in adult KA-ELS rats in the presence of an unfamiliar rat, supporting altered social anxiety following KA-ELS. Reanalysis of a social approach task further indicated abnormal social interactions. Taken together with previous physiological and behavioral data, these data support the hypothesis that KA-ELS lead to a latent autistic phenotype in adult rats not attributable to other early alterations in development.
Collapse
Affiliation(s)
- Paul B Bernard
- Department of Pediatrics, University of Colorado, School of Medicine, USA
| | - Anna M Castano
- Department of Pediatrics, University of Colorado, School of Medicine, USA
| | - Christy S Beitzel
- Department of Neuroscience Graduate Program, University of Colorado, School of Medicine, USA
| | - Vivian B Carlson
- Department of Pediatrics, University of Colorado, School of Medicine, USA
| | - Tim A Benke
- Department of Pediatrics, University of Colorado, School of Medicine, USA; Department of Neuroscience Graduate Program, University of Colorado, School of Medicine, USA; Department of Neurology, University of Colorado, School of Medicine, USA; Department of Pharmacology, University of Colorado, School of Medicine, USA; Department of Otolaryngology, University of Colorado, School of Medicine, USA.
| |
Collapse
|
32
|
Lozano R, Rosero CA, Hagerman RJ. Fragile X spectrum disorders. Intractable Rare Dis Res 2014; 3:134-46. [PMID: 25606363 PMCID: PMC4298643 DOI: 10.5582/irdr.2014.01022] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 11/28/2014] [Indexed: 12/13/2022] Open
Abstract
The fragile X mental retardation 1 gene (FMR1), which codes for the fragile X mental retardation 1 protein (FMRP), is located at Xp27.3. The normal allele of the FMR1 gene typically has 5 to 40 CGG repeats in the 5' untranslated region; abnormal alleles of dynamic mutations include the full mutation (> 200 CGG repeats), premutation (55-200 CGG repeats) and the gray zone mutation (45-54 CGG repeats). Premutation carriers are common in the general population with approximately 1 in 130-250 females and 1 in 250-810 males, whereas the full mutation and Fragile X syndrome (FXS) occur in approximately 1 in 4000 to 1 in 7000. FMR1 mutations account for a variety of phenotypes including the most common monogenetic cause of inherited intellectual disability (ID) and autism (FXS), the most common genetic form of ovarian failure, the fragile X-associated primary ovarian insufficiency (FXPOI, premutation); and fragile X-associated tremor/ataxia syndrome (FXTAS, premutation). The premutation can also cause developmental problems including ASD and ADHD especially in boys and psychopathology including anxiety and depression in children and adults. Some premutation carriers can have a deficit of FMRP and some unmethylated full mutation individuals can have elevated FMR1 mRNA that is considered a premutation problem. Therefore the term "Fragile X Spectrum Disorder" (FXSD) should be used to include the wide range of overlapping phenotypes observed in affected individuals with FMR1 mutations. In this review we focus on the phenotypes and genotypes of children with FXSD.
Collapse
Affiliation(s)
- Reymundo Lozano
- UC Davis MIND Institute and Department of Pediatrics, UC Davis Medical Center, Sacramento, CA, USA
- Address correspondence to: Dr. Reymundo Lozano, UC Davis MIND Institute and Department of Pediatrics, UC Davis Medical Center, Sacramento, CA, USA. E-mail:
| | - Carolina Alba Rosero
- Instituto Colombiano del Sistema Nervioso, Clínica Montserrat, Bogotá D.C, Colombia
| | - Randi J Hagerman
- UC Davis MIND Institute and Department of Pediatrics, UC Davis Medical Center, Sacramento, CA, USA
| |
Collapse
|
33
|
Bernard PB, Benke TA. Early life seizures: evidence for chronic deficits linked to autism and intellectual disability across species and models. Exp Neurol 2014; 263:72-8. [PMID: 25284323 DOI: 10.1016/j.expneurol.2014.09.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 09/02/2014] [Accepted: 09/16/2014] [Indexed: 11/08/2022]
Abstract
Recent work in Exp Neurol by Lugo et al. (2014b) demonstrated chronic alterations in sociability, learning and memory following multiple early life seizures (ELS) in a mouse model. This work adds to the growing body of evidence supporting the detrimental nature of ELS on the developing brain to contribute to aspects of an autistic phenotype with intellectual disability. Review of the face validity of behavioral testing and the construct validity of the models used informs the predictive ability and thus the utility of these models to translate underlying molecular and cellular mechanisms into future human studies.
Collapse
Affiliation(s)
- Paul B Bernard
- Department of Pediatrics, University of Colorado, School of Medicine, USA
| | - Tim A Benke
- Department of Pediatrics, University of Colorado, School of Medicine, USA; Neuroscience Graduate Program, University of Colorado, School of Medicine, USA; Department of Neurology, University of Colorado, School of Medicine, USA; Department of Pharmacology, University of Colorado, School of Medicine, USA; Department of Otolaryngology, University of Colorado, School of Medicine, USA.
| |
Collapse
|
34
|
Bartley CM, O’Keefe RA, Bordey A. FMRP S499 is phosphorylated independent of mTORC1-S6K1 activity. PLoS One 2014; 9:e96956. [PMID: 24806451 PMCID: PMC4013076 DOI: 10.1371/journal.pone.0096956] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 04/08/2014] [Indexed: 11/18/2022] Open
Abstract
Hyperactive mammalian target of rapamycin (mTOR) is associated with cognitive deficits in several neurological disorders including tuberous sclerosis complex (TSC). The phosphorylation of the mRNA-binding protein FMRP reportedly depends on mTOR complex 1 (mTORC1) activity via p70 S6 kinase 1 (S6K1). Because this phosphorylation is thought to regulate the translation of messages important for synaptic plasticity, we explored whether FMRP phosphorylation of the S6K1-dependent residue (S499) is altered in TSC and states of dysregulated TSC-mTORC1 signaling. Surprisingly, we found that FMRP S499 phosphorylation was unchanged in heterozygous and conditional Tsc1 knockout mice despite significantly elevated mTORC1-S6K1 activity. Neither up- nor down-regulation of the mTORC1-S6K1 axis in vivo or in vitro had any effect on phospho-FMRP S499 levels. In addition, FMRP S499 phosphorylation was unaltered in S6K1-knockout mice. Collectively, these data strongly suggest that FMRP S499 phosphorylation is independent of mTORC1-S6K1 activity and is not altered in TSC.
Collapse
Affiliation(s)
- Christopher M. Bartley
- Departments of Neurosurgery, and Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Medical Scientist Training Program, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Neurobiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Rachel A. O’Keefe
- Departments of Neurosurgery, and Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Angélique Bordey
- Departments of Neurosurgery, and Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
35
|
Bernard PB, Castano AM, Bayer KU, Benke TA. Necessary, but not sufficient: insights into the mechanisms of mGluR mediated long-term depression from a rat model of early life seizures. Neuropharmacology 2014; 84:1-12. [PMID: 24780380 DOI: 10.1016/j.neuropharm.2014.04.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 03/28/2014] [Accepted: 04/14/2014] [Indexed: 01/25/2023]
Abstract
Using the rat model of early life seizures (ELS), which has exaggerated mGluR mediated long-term depression of synaptic strength (mGluR-LTD) in adulthood, we probed the signaling cascades underlying mGluR-LTD induction. Several inhibitors completely blocked mGluR-LTD in control but not in ELS rats: the proteasome, the mammalian target of rapamycin (mTOR), S6 kinase (S6K), or L-type voltage-gated calcium channels (L-type VGCC). Inhibition of the Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) resulted in a near complete block of mGluR-LTD in control rats and a slight reduction of mGluR-LTD in ELS rats. "Autonomous" CaMKII was found to be upregulated in ELS rats, while elevated S6K activity, which is stimulated by mTOR, was described previously. Thus, modulation of each of these factors was necessary for mGluR-LTD induction in control rats, but even their combined, permanent activation in the ELS rats was not sufficient to individually support mGluR-LTD induction following ELS. This implies that while these factors may act sequentially in controls to mediate mGluR-LTD, this is no longer the case after ELS. In contrast, activated ERK was found to be significantly down-regulated in ELS rats. Inhibition of MEK/ERK activation in control rats elevated mGluR-LTD to the exaggerated levels seen in ELS rats. Together, these results elucidate both the mechanisms that persistently enhance mGluR-LTD after ELS and the mechanisms underlying normal mGluR-LTD by providing evidence for multiple, convergent pathways that mediate mGluR-LTD induction. With our prior work, this ties these signaling cascades to the ELS behavioral phenotype that includes abnormal working memory, fear conditioning and socialization.
Collapse
Affiliation(s)
- Paul B Bernard
- Department of Pediatrics, University of Colorado, School of Medicine, CO, USA
| | - Anna M Castano
- Department of Pediatrics, University of Colorado, School of Medicine, CO, USA
| | - K Ulrich Bayer
- Neuroscience Graduate Program, University of Colorado, School of Medicine, CO, USA; Department of Pharmacology, University of Colorado, School of Medicine, CO, USA
| | - Tim A Benke
- Department of Pediatrics, University of Colorado, School of Medicine, CO, USA; Neuroscience Graduate Program, University of Colorado, School of Medicine, CO, USA; Department of Neurology, University of Colorado, School of Medicine, CO, USA; Department of Pharmacology, University of Colorado, School of Medicine, CO, USA; Department of Otolaryngology, University of Colorado, School of Medicine, CO, USA.
| |
Collapse
|
36
|
Lugo JN, Swann JW, Anderson AE. Early-life seizures result in deficits in social behavior and learning. Exp Neurol 2014; 256:74-80. [PMID: 24685665 DOI: 10.1016/j.expneurol.2014.03.014] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 03/14/2014] [Accepted: 03/20/2014] [Indexed: 12/30/2022]
Abstract
Children with epilepsy show a high co-morbidity with psychiatric disorders and autism. One of the critical determinants of a child's behavioral outcome with autism and cognitive dysfunction is the age of onset of seizures. In order to examine whether seizures during postnatal days 7-11 result in learning and memory deficits and behavioral features of autism we administered the inhalant flurothyl to induce seizures in C57BL/6J mice. Mice received three seizures per day for five days starting on postnatal day 7. Parallel control groups consisted of similarly handled animals that were not exposed to flurothyl and naïve mice. Subjects were then processed through a battery of behavioral tests in adulthood: elevated-plus maze, nose-poke assay, marble burying, social partition, social chamber, fear conditioning, and Morris water maze. Mice with early-life seizures had learning and memory deficits in the training portion of the Morris water maze (p<0.05) and probe trial (p<0.01). Mice with seizures showed no differences in marble burying, the nose-poke assay, or elevated plus-maze testing compared to controls. However, they showed a significant difference in the social chamber and social partition tests. Mice with seizures during postnatal days 7-11 showed a significant decrease in social interaction in the social chamber test and had a significant impairment in social behavior in the social partition test. Together, these results indicate that early life seizures result in deficits in hippocampal-dependent memory tasks and produce long-term disruptions in social behavior.
Collapse
Affiliation(s)
- Joaquin N Lugo
- Department of Psychology and Neuroscience, Baylor University, Waco, TX 76798, USA; Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA.
| | - John W Swann
- Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Anne E Anderson
- Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
37
|
Polussa J, Schneider A, Hagerman R. Molecular Advances Leading to Treatment Implications for Fragile X Premutation Carriers. BRAIN DISORDERS & THERAPY 2014; 3:1000119. [PMID: 25436181 PMCID: PMC4245015 DOI: 10.4172/2168-975x.1000119] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Fragile X syndrome (FXS) is the most common single gene cause of intellectual disability and it is characterized by a CGG expansion of more than 200 repeats in the FMR1 gene, leading to methylation of the promoter and gene silencing. The fragile X premutation, characterized by a 55 to 200 CGG repeat expansion, causes health problems and developmental difficulties in some, but not all, carriers. The premutation causes primary ovarian insufficiency in approximately 20% of females, psychiatric problems (including depression and/or anxiety) in approximately 50% of carriers and a neurodegenerative disorder, the fragile X-associated tremor ataxia syndrome (FXTAS), in approximately 40% of males and 16% of females later in life. Recent clinical studies in premutation carriers have expanded the health problems that may be seen. Advances in the molecular pathogenesis of the premutation have shown significant mitochondrial dysfunction and oxidative stress in neurons which may be amenable to treatment. Here we review the clinical problems of carriers and treatment recommendations.
Collapse
Affiliation(s)
- Jonathan Polussa
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis Health System, Sacramento, California, USA
- Department of Pediatrics, University of California Davis Health System, Sacramento, California, USA
| | - Andrea Schneider
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis Health System, Sacramento, California, USA
- Department of Pediatrics, University of California Davis Health System, Sacramento, California, USA
| | - Randi Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis Health System, Sacramento, California, USA
- Department of Pediatrics, University of California Davis Health System, Sacramento, California, USA
| |
Collapse
|
38
|
Viscidi EW, Johnson AL, Spence SJ, Buka SL, Morrow EM, Triche EW. The association between epilepsy and autism symptoms and maladaptive behaviors in children with autism spectrum disorder. AUTISM : THE INTERNATIONAL JOURNAL OF RESEARCH AND PRACTICE 2013; 18:996-1006. [PMID: 24165273 DOI: 10.1177/1362361313508027] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Epilepsy is common in children with autism spectrum disorder (ASD) but little is known about how seizures impact the autism phenotype. The association between epilepsy and autism symptoms and associated maladaptive behaviors was examined in 2,645 children with ASD, of whom 139 had epilepsy, from the Simons Simplex Collection. Children with ASD and epilepsy had significantly more autism symptoms and maladaptive behaviors than children without epilepsy. However, after adjusting for IQ, only hyperactivity symptoms remained significantly increased (13% higher) in the epilepsy group. Among children with ASD without co-occurring intellectual disability, children with epilepsy had significantly more irritability (20% higher) and hyperactivity (24% higher) symptoms. This is the largest study to date comparing the autism phenotype in children with ASD with and without epilepsy. Children with ASD and epilepsy showed greater impairment than children without epilepsy, which was mostly explained by the lower IQ of the epilepsy group. These findings have important clinical implications for patients with ASD.
Collapse
Affiliation(s)
| | - Ashley L Johnson
- Emma Pendleton Bradley Hospital, USA Brown University Medical School, USA
| | | | | | - Eric M Morrow
- Brown University School of Public Health, USA Emma Pendleton Bradley Hospital, USA Brown University Medical School, USA
| | | |
Collapse
|