1
|
Reyes-Lizaola S, Luna-Zarate U, Tendilla-Beltrán H, Morales-Medina JC, Flores G. Structural and biochemical alterations in dendritic spines as key mechanisms for severe mental illnesses. Prog Neuropsychopharmacol Biol Psychiatry 2024; 129:110876. [PMID: 37863171 DOI: 10.1016/j.pnpbp.2023.110876] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/22/2023]
Abstract
Severe mental illnesses (SMI) collectively affect approximately 20% of the global population, as estimated by the World Health Organization (WHO). Despite having diverse etiologies, clinical symptoms, and pharmacotherapies, these diseases share a common pathophysiological characteristic: the misconnection of brain areas involved in reality perception, executive control, and cognition, including the corticolimbic system. Dendritic spines play a crucial role in excitatory neurotransmission within the central nervous system. These small structures exhibit remarkable plasticity, regulated by factors such as neurotransmitter tone, neurotrophic factors, and innate immunity-related molecules, and other mechanisms - all of which are associated with the pathophysiology of SMI. However, studying dendritic spine mechanisms in both healthy and pathological conditions in patients is fraught with technical limitations. This is where animal models related to these diseases become indispensable. They have played a pivotal role in elucidating the significance of dendritic spines in SMI. In this review, the information regarding the potential role of dendritic spines in SMI was summarized, drawing from clinical and animal model reports. Also, the implications of targeting dendritic spine-related molecules for SMI treatment were explored. Specifically, our focus is on major depressive disorder and the neurodevelopmental disorders schizophrenia and autism spectrum disorder. Abundant clinical and basic research has studied the functional and structural plasticity of dendritic spines in these diseases, along with potential pharmacological targets that modulate the dynamics of these structures. These targets may be associated with the clinical efficacy of the pharmacotherapy.
Collapse
Affiliation(s)
- Sebastian Reyes-Lizaola
- Departamento de Ciencias de la Salud, Licenciatura en Medicina, Universidad Popular del Estado de Puebla (UPAEP), Puebla, Mexico
| | - Ulises Luna-Zarate
- Departamento de Ciencias de la Salud, Licenciatura en Medicina, Universidad de las Américas Puebla (UDLAP), Puebla, Mexico
| | - Hiram Tendilla-Beltrán
- Laboratorio de Neuropsiquiatría, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Julio César Morales-Medina
- Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | - Gonzalo Flores
- Laboratorio de Neuropsiquiatría, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico.
| |
Collapse
|
2
|
Baker MR, Lee AS, Rajadhyaksha AM. L-type calcium channels and neuropsychiatric diseases: Insights into genetic risk variant-associated genomic regulation and impact on brain development. Channels (Austin) 2023; 17:2176984. [PMID: 36803254 PMCID: PMC9980663 DOI: 10.1080/19336950.2023.2176984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 02/01/2023] [Indexed: 02/21/2023] Open
Abstract
Recent human genetic studies have linked a variety of genetic variants in the CACNA1C and CACNA1D genes to neuropsychiatric and neurodevelopmental disorders. This is not surprising given the work from multiple laboratories using cell and animal models that have established that Cav1.2 and Cav1.3 L-type calcium channels (LTCCs), encoded by CACNA1C and CACNA1D, respectively, play a key role in various neuronal processes that are essential for normal brain development, connectivity, and experience-dependent plasticity. Of the multiple genetic aberrations reported, genome-wide association studies (GWASs) have identified multiple single nucleotide polymorphisms (SNPs) in CACNA1C and CACNA1D that are present within introns, in accordance with the growing body of literature establishing that large numbers of SNPs associated with complex diseases, including neuropsychiatric disorders, are present within non-coding regions. How these intronic SNPs affect gene expression has remained a question. Here, we review recent studies that are beginning to shed light on how neuropsychiatric-linked non-coding genetic variants can impact gene expression via regulation at the genomic and chromatin levels. We additionally review recent studies that are uncovering how altered calcium signaling through LTCCs impact some of the neuronal developmental processes, such as neurogenesis, neuron migration, and neuron differentiation. Together, the described changes in genomic regulation and disruptions in neurodevelopment provide possible mechanisms by which genetic variants of LTCC genes contribute to neuropsychiatric and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Madelyn R. Baker
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, USA
| | - Andrew S. Lee
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, USA
- Developmental Biology Program, Sloan Kettering Institute, New York, USA
| | - Anjali M. Rajadhyaksha
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, USA
- Pediatric Neurology, Department of Pediatrics, Weill Cornell Medicine, New York, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, USA
- Weill Cornell Autism Research Program, Weill Cornell Medicine, New York, USA
| |
Collapse
|
3
|
Li SY, Zhao X, Cheng MY, Lu L, Guo JX, Xuan DS, Sun YB, Xing QN, Meng LS, Liao JJ, Cui SH, Zhang LJ, Feng ZQ, Zhang XA. Quantitative Relaxometry Assessment of Brain Microstructural Abnormality of Preschool Children With Autism Spectrum Disorder With Synthetic Magnetic Resonance Imaging. J Comput Assist Tomogr 2023; 47:959-966. [PMID: 37948372 DOI: 10.1097/rct.0000000000001507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
OBJECTIVE This study aimed to perform an assessment of brain microstructure in children with autism aged 2 to 5 years using relaxation times acquired by synthetic magnetic resonance imaging. MATERIALS AND METHODS Thirty-four children with autism spectrum disorder (ASD) (ASD group) and 17 children with global developmental delay (GDD) (GDD group) were enrolled, and synthetic magnetic resonance imaging was performed to obtain T1 and T2 relaxation times. The differences in brain relaxation times between the 2 groups of children were compared, and the correlation between significantly changed T1/T2 and clinical neuropsychological scores in the ASD group was analyzed. RESULTS Compared with the GDD group, shortened T1 relaxation times in the ASD group were distributed in the genu of corpus callosum (GCC) ( P = 0.003), splenium of corpus callosum ( P = 0.002), and right thalamus (TH) ( P = 0.014), whereas shortened T2 relaxation times in the ASD group were distributed in GCC ( P = 0.011), left parietal white matter ( P = 0.035), and bilateral TH (right, P = 0.014; left, P = 0.016). In the ASD group, the T2 of the left parietal white matter is positively correlated with gross motor (developmental quotient [DQ] 2) and personal-social behavior (DQ5), respectively ( r = 0.377, P = 0.028; r = 0.392, P = 0.022); the T2 of the GCC was positively correlated with DQ5 ( r = 0.404, P = 0.018); and the T2 of the left TH is positively correlated with DQ2 and DQ5, respectively ( r = 0.433, P = 0.009; r = 0.377, P = 0.028). All significantly changed relaxation values were not significantly correlated with Childhood Autism Rating Scale scores. CONCLUSIONS The shortened relaxometry times in the brain of children with ASD may be associated with the increased myelin content and decreased water content in the brain of children with ASD in comparison with GDD, contributing the understanding of the pathophysiology of ASD. Therefore, the T1 and T2 relaxometry may be used as promising imaging markers for ASD diagnosis.
Collapse
Affiliation(s)
- Shuang-Yu Li
- From the Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - Xin Zhao
- From the Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - Mei-Ying Cheng
- From the Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - Lin Lu
- From the Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou
| | | | - De-Sheng Xuan
- From the Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - Yong-Bing Sun
- Zhengzhou University People's Hospital, Zhengzhou, China
| | - Qing-Na Xing
- From the Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - Ling-Song Meng
- From the Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - Jun-Jie Liao
- From the Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - Shu-Hong Cui
- From the Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - Ling-Jie Zhang
- From the Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - Zhan-Qi Feng
- From the Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - Xiao-An Zhang
- From the Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou
| |
Collapse
|
4
|
Gorlova A, Svirin E, Pavlov D, Cespuglio R, Proshin A, Schroeter CA, Lesch KP, Strekalova T. Understanding the Role of Oxidative Stress, Neuroinflammation and Abnormal Myelination in Excessive Aggression Associated with Depression: Recent Input from Mechanistic Studies. Int J Mol Sci 2023; 24:915. [PMID: 36674429 PMCID: PMC9861430 DOI: 10.3390/ijms24020915] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/26/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Aggression and deficient cognitive control problems are widespread in psychiatric disorders, including major depressive disorder (MDD). These abnormalities are known to contribute significantly to the accompanying functional impairment and the global burden of disease. Progress in the development of targeted treatments of excessive aggression and accompanying symptoms has been limited, and there exists a major unmet need to develop more efficacious treatments for depressed patients. Due to the complex nature and the clinical heterogeneity of MDD and the lack of precise knowledge regarding its pathophysiology, effective management is challenging. Nonetheless, the aetiology and pathophysiology of MDD has been the subject of extensive research and there is a vast body of the latest literature that points to new mechanisms for this disorder. Here, we overview the key mechanisms, which include neuroinflammation, oxidative stress, insulin receptor signalling and abnormal myelination. We discuss the hypotheses that have been proposed to unify these processes, as many of these pathways are integrated for the neurobiology of MDD. We also describe the current translational approaches in modelling depression, including the recent advances in stress models of MDD, and emerging novel therapies, including novel approaches to management of excessive aggression, such as anti-diabetic drugs, antioxidant treatment and herbal compositions.
Collapse
Affiliation(s)
- Anna Gorlova
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Laboratory of Cognitive Dysfunctions, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
| | - Evgeniy Svirin
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Laboratory of Cognitive Dysfunctions, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
- Neuroplast BV, 6222 NK Maastricht, The Netherlands
| | - Dmitrii Pavlov
- Hotchkiss Brain Institute, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Raymond Cespuglio
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Centre de Recherche en Neurosciences de Lyon (CRNL), 69500 Bron, France
| | - Andrey Proshin
- P.K. Anokhin Research Institute of Normal Physiology, 125315 Moscow, Russia
| | - Careen A. Schroeter
- Preventive and Environmental Medicine, Kastanienhof Clinic, 50858 Köln-Junkersdorf, Germany
| | - Klaus-Peter Lesch
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, The Netherlands
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Tatyana Strekalova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, The Netherlands
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
5
|
Aberrant Ganglioside Functions to Underpin Dysregulated Myelination, Insulin Signalling, and Cytokine Expression: Is There a Link and a Room for Therapy? Biomolecules 2022; 12:biom12101434. [PMID: 36291644 PMCID: PMC9599472 DOI: 10.3390/biom12101434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/17/2022] Open
Abstract
Gangliosides are molecules widely present in the plasma membranes of mammalian cells, participating in a variety of processes, including protein organization, transmembrane signalling and cell adhesion. Gangliosides are abundant in the grey matter of the brain, where they are critically involved in postnatal neural development and function. The common precursor of the majority of brain gangliosides, GM3, is formed by the sialylation of lactosylceramide, and four derivatives of its a- and b-series, GM1, GD1a, GD1b and GT1b, constitute 95% of all the brain gangliosides. Impairments in ganglioside metabolism due to genetic abnormalities of GM-synthases are associated with severe neurological disorders. Apart from that, the latest genome-wide association and translational studies suggest a role of genes involved in brain ganglioside synthesis in less pervasive psychiatric disorders. Remarkably, the most recent animal studies showed that abnormal ganglioside functions result in dysregulated neuroinflammation, aberrant myelination and altered insulin receptor signalling. At the same time, these molecular features are well established as accompanying developmental psychiatric disorders such as attention-deficit hyperactivity disorder (ADHD) and autism spectrum disorders (ASD). This led us to hypothesize a role of deficient ganglioside function in developmental neuropsychiatric disorders and warrants further gene association clinical studies addressing this question. Here, we critically review the literature to discuss this hypothesis and focus on the recent studies on ST3GAL5-deficient mice. In addition, we elaborate on the therapeutic potential of various anti-inflammatory remedies for treatment of developmental neuropsychiatric conditions related to aberrant ganglioside functions.
Collapse
|
6
|
Malara M, Lutz AK, Incearap B, Bauer HF, Cursano S, Volbracht K, Lerner JJ, Pandey R, Delling JP, Ioannidis V, Arévalo AP, von Bernhardi JE, Schön M, Bockmann J, Dimou L, Boeckers TM. SHANK3 deficiency leads to myelin defects in the central and peripheral nervous system. Cell Mol Life Sci 2022; 79:371. [PMID: 35726031 PMCID: PMC9209365 DOI: 10.1007/s00018-022-04400-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/11/2022] [Accepted: 05/25/2022] [Indexed: 01/04/2023]
Abstract
Mutations or deletions of the SHANK3 gene are causative for Phelan–McDermid syndrome (PMDS), a syndromic form of autism spectrum disorders (ASDs). We analyzed Shank3Δ11(−/−) mice and organoids from PMDS individuals to study effects on myelin. SHANK3 was found to be expressed in oligodendrocytes and Schwann cells, and MRI analysis of Shank3Δ11(−/−) mice revealed a reduced volume of the corpus callosum as seen in PMDS patients. Myelin proteins including myelin basic protein showed significant temporal and regional differences with lower levels in the CNS but increased amounts in the PNS of Shank3Δ11(−/−) animals. Node, as well as paranode, lengths were increased and ultrastructural analysis revealed region-specific alterations of the myelin sheaths. In PMDS hiPSC-derived cerebral organoids we observed an altered number and delayed maturation of myelinating cells. These findings provide evidence that, in addition to a synaptic deregulation, impairment of myelin might profoundly contribute to the clinical manifestation of SHANK3 deficiency.
Collapse
Affiliation(s)
- Mariagiovanna Malara
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
- International Graduate School in Molecular Medicine, IGradU, 89081, Ulm, Germany
| | - Anne-Kathrin Lutz
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Berra Incearap
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
- International Graduate School in Molecular Medicine, IGradU, 89081, Ulm, Germany
| | - Helen Friedericke Bauer
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
- International Graduate School in Molecular Medicine, IGradU, 89081, Ulm, Germany
| | - Silvia Cursano
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Katrin Volbracht
- Molecular and Translational Neuroscience, Department of Neurology, Ulm University, 89081, Ulm, Germany
| | - Joanna Janina Lerner
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
- International Graduate School in Molecular Medicine, IGradU, 89081, Ulm, Germany
| | - Rakshita Pandey
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Jan Philipp Delling
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Valentin Ioannidis
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Andrea Pérez Arévalo
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | | | - Michael Schön
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Jürgen Bockmann
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Leda Dimou
- Molecular and Translational Neuroscience, Department of Neurology, Ulm University, 89081, Ulm, Germany
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany.
- DZNE, Ulm Site, 89081, Ulm, Germany.
| |
Collapse
|
7
|
Roberts TPL, Bloy L, Ku M, Blaskey L, Jackel CR, Edgar JC, Berman JI. A Multimodal Study of the Contributions of Conduction Velocity to the Auditory Evoked Neuromagnetic Response: Anomalies in Autism Spectrum Disorder. Autism Res 2020; 13:1730-1745. [PMID: 32924333 DOI: 10.1002/aur.2369] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 06/22/2020] [Accepted: 06/26/2020] [Indexed: 11/12/2022]
Abstract
This multimodal imaging study used magnetoencephalography, diffusion magnetic resonance imaging (MRI), and gamma-aminobutyric acid (GABA) magnetic resonance spectroscopy (MRS) to identify and contrast the multiple physiological mechanisms associated with auditory processing efficiency in typically developing (TD) children and children with autism spectrum disorder (ASD). Efficient transmission of auditory input between the ear and auditory cortex is necessary for rapid encoding of auditory sensory information. It was hypothesized that the M50 auditory evoked response latency would be modulated by white matter microstructure (indexed by diffusion MRI) and by tonic inhibition (indexed by GABA MRS). Participants were 77 children diagnosed with ASD and 40 TD controls aged 7-17 years. A model of M50 latency with auditory radiation fractional anisotropy and age as independent variables was able to predict 52% of M50 latency variance in TD children, but only 12% of variance in ASD. The ASD group exhibited altered patterns of M50 latency modulation characterized by both higher variance and deviation from the expected structure-function relationship established with the TD group. The TD M50 latency model was used to identify a subpopulation of ASD who are significant "outliers" to the TD model. The ASD outlier group exhibited unexpectedly long M50 latencies in conjunction with significantly lower GABA levels. These findings indicate the dependence of electrophysiologic sensory response latency on underlying microstructure (white matter) and neurochemistry (synaptic activity). This study demonstrates the use of biologically based measures to stratify ASD according to their brain-level "building blocks" as an alternative to their behavioral phenotype. LAY SUMMARY: Children with ASD often have a slower brain response when hearing sounds. This study used multiple brain imaging techniques to examine the structural and neurochemical factors which control the brain's response time to auditory tones in children with ASD and TD children. The relationship between brain imaging measures and brain response time was also used to identify ASD subgroups. Autism Res 2020, 13: 1730-1745. © 2020 International Society for Autism Research and Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Timothy P L Roberts
- Department of Radiology, Lurie Family Foundations MEG Imaging Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Luke Bloy
- Department of Radiology, Lurie Family Foundations MEG Imaging Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Matt Ku
- Department of Radiology, Lurie Family Foundations MEG Imaging Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Lisa Blaskey
- Department of Radiology, Lurie Family Foundations MEG Imaging Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Child and Adolescent Psychiatry and Behavioral Sciences, Center for Autism Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Carissa R Jackel
- Department of Radiology, Lurie Family Foundations MEG Imaging Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Division of Developmental and Behavioral Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - James Christopher Edgar
- Department of Radiology, Lurie Family Foundations MEG Imaging Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jeffrey I Berman
- Department of Radiology, Lurie Family Foundations MEG Imaging Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Ruigrok ANV, Lai MC. Sex/gender differences in neurology and psychiatry: Autism. HANDBOOK OF CLINICAL NEUROLOGY 2020; 175:283-297. [PMID: 33008532 DOI: 10.1016/b978-0-444-64123-6.00020-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Autism is a heterogenous set of early-onset neurodevelopmental conditions that are more prevalent in males than in females. Due to the high phenotypic, neurobiological, developmental, and etiological heterogeneity in the autism spectrum, recent research programs are increasingly exploring whether sex- and gender-related factors could be helpful markers to clarify the heterogeneity in autism and work toward a personalized approach to intervention and support. In this chapter, we summarize recent clinical and neuroscientific research addressing sex/gender influences in autism and explore how sex/gender-based investigations shed light on similar or different underlying neurodevelopmental mechanisms of autism by sex/gender. We review evidence that may help to explain some of the underlying sex-related biological mechanisms associated with autism, including genetics and the effects of sex steroid hormones in the prenatal environment. We conclude that current research points toward coexisting quantitative and, perhaps more evidently, qualitative sex/gender-modulation effects in autism across multiple neurobiological aspects. However, converging findings of specific neurobiological presentations and sex/gender-informed mechanisms cutting across the many subgroups within the autism spectrum are still lacking. Future research should use big data approaches and new stratification methods to decompose sex/gender-related heterogeneity in autism and work toward personalized, sex/gender-informed intervention and support for autistic people.
Collapse
Affiliation(s)
- Amber N V Ruigrok
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Meng-Chuan Lai
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom; Centre for Addiction and Mental Health & The Hospital for Sick Children, Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
9
|
Hirayama T, Hiraoka Y, Kitamura E, Miyazaki S, Horie K, Fukuda T, Hidema S, Koike M, Itakura A, Takeda S, Nishimori K. Oxytocin induced labor causes region and sex-specific transient oligodendrocyte cell death in neonatal mouse brain. J Obstet Gynaecol Res 2019; 46:66-78. [PMID: 31746074 DOI: 10.1111/jog.14149] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 10/07/2019] [Indexed: 12/16/2022]
Abstract
AIM Previous reports showed associations between oxytocin induced labor and mental disorders in offspring. However, those reports are restricted in epidemiological analyses and its mechanism remains unclear. In this study, we hypothesized that induced labor directly causes brain damage in newborns and results in the development of mental disorders. Therefore we aimed to investigate this hypothesis with animal model. METHODS The animal model of induced labor was established by subcutaneous oxytocin administration to term-pregnant C57BL/6J mice. We investigated the neonatal brain damage with evaluating immediate early gene expression (c-Fos, c-Jun and JunB) by quantitative polymerase reaction and TdT-mediated dUTP nick end labeling staining. To investigate the injured brain cell types, we performed double-immunostaining with TdT-mediated dUTP nick end labeling staining and each brain component specific protein, such as Oligo2, NeuN, GFAP and Iba1. RESULTS Brain damage during induced labor led to cell death in specific brain regions, which are implicated in mental disorders, in only male offspring at P0. Furthermore, oligodendrocyte precursors were selectively vulnerable compared to the other cell types. This oligodendrocyte-specific impairment during the perinatal period led to an increased numbers of Olig2-positive cells at P5. Expression levels of oxytocin and Oxtr in the fetal brain were not affected by the oxytocin administered to mothers during induced labor. CONCLUSION Oligodendrocyte cell death in specific brain regions, which was unrelated to the oxytocin itself, was caused by induced labor in only male offspring. This may be an underlying mechanism explaining the human epidemiological data suggesting an association between induced labor and mental disorders.
Collapse
Affiliation(s)
- Takashi Hirayama
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Department of Obstetrics and Gynecology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yuichi Hiraoka
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Laboratory for Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Eri Kitamura
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shinji Miyazaki
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Kengo Horie
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Tomokazu Fukuda
- Laboratory of Cell Engineering and Molecular Genetics, Iwate University Faculty of Science and Engineering, Morioka, Japan
| | - Shizu Hidema
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Masato Koike
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Atsuo Itakura
- Department of Obstetrics and Gynecology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Satoru Takeda
- Department of Obstetrics and Gynecology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | | |
Collapse
|
10
|
Abstract
Autism spectrum disorder (ASD) has been hypothesized to be a result of altered connectivity in the brain. Recent imaging studies suggest accelerated maturation of the white matter in young children with ASD, with underlying mechanisms unknown. Myelin is an integral part of the white matter and critical for connectivity; however, its role in ASD remains largely unclear. Here, we investigated myelin development in a model of idiopathic ASD, the BTBR mice. Magnetic resonance imaging revealed that fiber tracts in the frontal brain of the BTBR mice had increased volume at postnatal day 6, but the difference reduced over time, reminiscent of the findings in young patients. We further identified that myelination in the frontal brain of both male and female neonatal BTBR mice was increased, associated with elevated levels of myelin basic protein. However, myelin pattern was unaltered in adult BTBR mice, revealing accelerated developmental trajectory of myelination. Consistently, we found that signaling of platelet-derived growth factor receptor alpha (PDGFRα) was reduced in the frontal brain of neonatal BTBR mice. However, levels of microRNA species known to regulate PDGFRα signaling and myelination were unaltered. Together, these results suggest that precocious myelination could potentially contribute to increased volume and connectivity of the white matter observed in young children with ASD.
Collapse
|
11
|
Volpon Santos M, da Silva Lopes L, Machado HR, Santos de Oliveira R. Behavioral and Biochemical Features of the Course and Surgical Treatment of Experimental Obstructive Hydrocephalus in Young Rats. Dev Neurosci 2019; 41:34-43. [PMID: 30999305 DOI: 10.1159/000497433] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 02/01/2019] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Hydrocephalus is a multifactorial disease, affecting the dynamics of cerebrospinal fluid (CSF) and leading to severe neurological impairment in children; in spite of the recent advances in hydrocephalus research, it has many physiopathological aspects that still remain poorly understood, especially after treatment. OBJECTIVES To analyze the clinical, radiological, histopathological, and biochemical aspects of kaolin-induced hydrocephalus in an experimental model, both in the acute phase and after shunt treatment, by means of behavioral tests, magnetic resonance imaging (MRI) scans, histopathological studies, and level of inflammatory interleukins in the CSF. METHODS Seven-day-old Wistar rats were used and subdivided into three subgroups: treated hydrocephalic (n = 24), untreated hydrocephalic (n = 17), and controls (n = 5). The hydrocephalic groups underwent cisternal injection of 15% kaolin for induction of hydrocephalus at 7 days of age. The treated group was submitted to a ventricular-subcutaneous shunt (VSCS) 1 week after induction. All animals were euthanized at 21 days of age. They underwent motor function and memory testing as well as brain MRI scans. Histopathological analysis for glial fibrillary acidic protein and Ki-67 was done, and CSF was collected for measurement of IL-1β, IL-6, and TNF-α. RESULTS The average time to reach the water maze platform was highest in the untreated hydrocephalic group. The magnetization transfer rates were 37.21 and 33.76 before and after shunting, respectively. The mean astrocyte counts were 2.45, 1.36, and 90.5 for shunted, untreated, and control animals, respectively. The mean CSF IL-1β concentrations were 62.3 and 249.6 pg/mL, the average IL-6 levels were 104.2 and 364.7 pg/mL, and the average TNF-α values were 4.9 and 170.5 pg/mL for the treated hydrocephalic group and the untreated group, respectively. CONCLUSIONS Pups treated with a CSF shunt showed better performance on memory tests. VSCS did not revert demyelination caused by hydrocephalus. Likewise, reactive astrocytosis and cell proliferation over the germinal matrix were not reversed after shunting. Hydrocephalic animals had raised levels of inflammatory interleukins, which returned to normal after treatment.
Collapse
Affiliation(s)
- Marcelo Volpon Santos
- Division of Pediatric Neurosurgery, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil, .,Developmental Neuropathology Laboratory, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil,
| | - Luiza da Silva Lopes
- Developmental Neuropathology Laboratory, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Hélio Rubens Machado
- Division of Pediatric Neurosurgery, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Ricardo Santos de Oliveira
- Division of Pediatric Neurosurgery, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
12
|
Carter CJ. Autism genes and the leukocyte transcriptome in autistic toddlers relate to pathogen interactomes, infection and the immune system. A role for excess neurotrophic sAPPα and reduced antimicrobial Aβ. Neurochem Int 2019; 126:36-58. [PMID: 30862493 DOI: 10.1016/j.neuint.2019.03.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 02/22/2019] [Accepted: 03/06/2019] [Indexed: 12/20/2022]
Abstract
Prenatal and early childhood infections have been implicated in autism. Many autism susceptibility genes (206 Autworks genes) are localised in the immune system and are related to immune/infection pathways. They are enriched in the host/pathogen interactomes of 18 separate microbes (bacteria/viruses and fungi) and to the genes regulated by bacterial toxins, mycotoxins and Toll-like receptor ligands. This enrichment was also observed for misregulated genes from a microarray study of leukocytes from autistic toddlers. The upregulated genes from this leukocyte study also matched the expression profiles in response to numerous infectious agents from the Broad Institute molecular signatures database. They also matched genes related to sudden infant death syndrome and autism comorbid conditions (autoimmune disease, systemic lupus erythematosus, diabetes, epilepsy and cardiomyopathy) as well as to estrogen and thyrotropin responses and to those upregulated by different types of stressors including oxidative stress, hypoxia, endoplasmic reticulum stress, ultraviolet radiation or 2,4-dinitrofluorobenzene, a hapten used to develop allergic skin reactions in animal models. The oxidative/integrated stress response is also upregulated in the autism brain and may contribute to myelination problems. There was also a marked similarity between the expression signatures of autism and Alzheimer's disease, and 44 shared autism/Alzheimer's disease genes are almost exclusively expressed in the blood-brain barrier. However, in contrast to Alzheimer's disease, levels of the antimicrobial peptide beta-amyloid are decreased and the levels of the neurotrophic/myelinotrophic soluble APP alpha are increased in autism, together with an increased activity of α-secretase. sAPPα induces an increase in glutamatergic and a decrease in GABA-ergic synapses creating and excitatory/inhibitory imbalance that has also been observed in autism. A literature survey showed that multiple autism genes converge on APP processing and that many are able to increase sAPPalpha at the expense of beta-amyloid production. A genetically programmed tilt of this axis towards an overproduction of neurotrophic/gliotrophic sAPPalpha and underproduction of antimicrobial beta-amyloid may explain the brain overgrowth and myelination dysfunction, as well as the involvement of pathogens in autism.
Collapse
Affiliation(s)
- C J Carter
- PolygenicPathways, 41C Marina, Saint Leonard's on Sea, TN38 0BU, East Sussex, UK.
| |
Collapse
|
13
|
Sui YV, Donaldson J, Miles L, Babb JS, Castellanos FX, Lazar M. Diffusional kurtosis imaging of the corpus callosum in autism. Mol Autism 2018; 9:62. [PMID: 30559954 PMCID: PMC6293510 DOI: 10.1186/s13229-018-0245-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 11/20/2018] [Indexed: 12/31/2022] Open
Abstract
Background The corpus callosum is implicated in the pathophysiology of autism spectrum disorder (ASD). However, specific structural deficits and underlying mechanisms are yet to be well defined. Methods We employed diffusional kurtosis imaging (DKI) metrics to characterize white matter properties within five discrete segments of the corpus callosum in 17 typically developing (TD) adults and 16 age-matched participants with ASD without co-occurring intellectual disability (ID). The DKI metrics included axonal water fraction (faxon) and intra-axonal diffusivity (Daxon), which reflect axonal density and caliber, and extra-axonal radial (RDextra) and axial (ADextra) diffusivities, which reflect myelination and microstructural organization of the extracellular space. The relationships between DKI metrics and processing speed, a cognitive feature known to be impaired in ASD, were also examined. Results ASD group had significantly decreased callosal faxon and Daxon (p = .01 and p = .045), particularly in the midbody, isthmus, and splenium. Regression analysis showed that variation in DKI metrics, primarily in the mid and posterior callosal regions explained up to 70.7% of the variance in processing speed scores for TD (p = .001) but not for ASD (p > .05). Conclusion Decreased DKI metrics suggested that ASD may be associated with axonal deficits such as reduced axonal caliber and density in the corpus callosum, especially in the mid and posterior callosal areas. These data suggest that impaired interhemispheric connectivity may contribute to decreased processing speed in ASD participants. Electronic supplementary material The online version of this article (10.1186/s13229-018-0245-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yu Veronica Sui
- 1Department of Radiology, New York University School of Medicine, New York, NY USA.,4Center for Biomedical Imaging, NYU Langone Health, 660 First Ave, 4th floor, New York, NY 10016 USA
| | - Jeffrey Donaldson
- 1Department of Radiology, New York University School of Medicine, New York, NY USA
| | - Laura Miles
- 1Department of Radiology, New York University School of Medicine, New York, NY USA
| | - James S Babb
- 1Department of Radiology, New York University School of Medicine, New York, NY USA
| | - Francisco Xavier Castellanos
- 2Department of Child and Adolescent Psychiatry, Hassenfeld Children's Hospital at NYU Langone, New York, NY USA.,3Nathan Kline Institute for Psychiatric Research, Orangeburg, NY USA
| | - Mariana Lazar
- 1Department of Radiology, New York University School of Medicine, New York, NY USA.,4Center for Biomedical Imaging, NYU Langone Health, 660 First Ave, 4th floor, New York, NY 10016 USA
| |
Collapse
|
14
|
Cheli VT, Santiago González DA, Zamora NN, Lama TN, Spreuer V, Rasmusson RL, Bett GC, Panagiotakos G, Paez PM. Enhanced oligodendrocyte maturation and myelination in a mouse model of Timothy syndrome. Glia 2018; 66:2324-2339. [PMID: 30151840 PMCID: PMC6697123 DOI: 10.1002/glia.23468] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 12/13/2017] [Accepted: 05/16/2018] [Indexed: 01/09/2023]
Abstract
To study the role of L-type voltage-gated Ca++ channels in oligodendrocyte development, we used a mouse model of Timothy syndrome (TS) in which a gain-of-function mutation in the α1 subunit of the L-type Ca++ channel Cav1.2 gives rise to an autism spectrum disorder (ASD). Oligodendrocyte progenitor cells (OPCs) isolated from the cortex of TS mice showed greater L-type Ca++ influx and displayed characteristics suggestive of advanced maturation compared to control OPCs, including a more complex morphology and higher levels of myelin protein expression. Consistent with this, expression of Cav1.2 channels bearing the TS mutation in wild-type OPCs triggered process formation and promoted oligodendrocyte-neuron interaction via the activation of Ca++ /calmodulin-dependent protein kinase II. To ascertain whether accelerated OPC maturation correlated with functional enhancements, we examined myelination in the TS brain at different postnatal time points. The expression of myelin proteins was significantly higher in the corpus callosum, cortex and striatum of TS animals, and immunohistochemical analysis for oligodendrocyte stage-specific markers revealed an increase in the density of myelinating oligodendrocytes in several areas of the TS brain. Along the same line, electron microscopy studies in the corpus callosum of TS animals showed significant increases both in the percentage of myelinated axons and in the thickness of myelin sheaths. In summary, these data indicate that OPC development and oligodendrocyte myelination is enhanced in the brain of TS mice, and suggest that this mouse model of a syndromic ASD is a useful tool to explore the role of L-type Ca++ channels in myelination.
Collapse
Affiliation(s)
- Veronica T. Cheli
- Hunter James Kelly Research Institute, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York
| | - Diara A. Santiago González
- Hunter James Kelly Research Institute, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York
| | - Norma N. Zamora
- Hunter James Kelly Research Institute, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York
| | - Tenzing N. Lama
- Hunter James Kelly Research Institute, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York
| | - Vilma Spreuer
- Hunter James Kelly Research Institute, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York
| | - Randall L. Rasmusson
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York
| | - Glenna C. Bett
- Department of Obstetrics and Gynecology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York
| | - Georgia Panagiotakos
- Department of Biochemistry and Biophysics and Kavli Institute for Fundamental Neuroscience, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, School of Medicine, University of California at San Francisco, San Francisco, California
| | - Pablo M. Paez
- Hunter James Kelly Research Institute, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York
| |
Collapse
|
15
|
Dickinson A, DiStefano C, Lin YY, Scheffler AW, Senturk D, Jeste SS. Interhemispheric alpha-band hypoconnectivity in children with autism spectrum disorder. Behav Brain Res 2018; 348:227-234. [PMID: 29689375 PMCID: PMC5993636 DOI: 10.1016/j.bbr.2018.04.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 04/12/2018] [Accepted: 04/17/2018] [Indexed: 12/16/2022]
Abstract
Diverse genetic and environmental etiologies converge onto circuit level brain dysfunction in autism spectrum disorder (ASD), manifesting at a macroscopic level as aberrant neural connectivity. Previous studies have described atypical patterns of decreased short range and increased long range connectivity in ASD [1 ]. However, it remains unclear whether group level features of circuit dysfunction are consistently present across the range of cognitive function seen in the autism spectrum. The dynamics of neural oscillations in the alpha range (6-12 Hz) are exquisitely sensitive to healthy development of functional and structural connectivity. Alpha-band coherence, measured with high temporal-precision electroencephalography (EEG) therefore represents an ideal tool for studying neural connectivity in developmental populations. Here we examined spontaneous alpha phase coherence in a heterogeneous sample of 59 children with ASD and 39 age matched typically developing children. Using a data driven approach, we conducted an unbiased examination of all possible atypical connectivity patterns across all cortical regions. Long-range hypoconnectivity was present in children with ASD compared to typically developing children, with temporal interhemispheric connectivity showing the largest difference between the two groups. Decreased long range alpha coherence distinguishes a heterogeneous group of ASD children from typically developing children. Interhemispheric temporal hypoconnectivity represents a fundamental functional difference in children with ASD across a wide cognitive and age range that may reflect white matter disturbances or increased signal variability at temporal sites in ASD.
Collapse
Affiliation(s)
- Abigail Dickinson
- Center for Autism Research and Treatment, University of California, Semel Institute for Neuroscience, 760 Westwood Plaza, Suite A7-452 Los Angeles, CA, 90095, United States.
| | - Charlotte DiStefano
- Center for Autism Research and Treatment, University of California, Semel Institute for Neuroscience, 760 Westwood Plaza, Suite A7-452 Los Angeles, CA, 90095, United States
| | - Yin-Ying Lin
- Center for Autism Research and Treatment, University of California, Semel Institute for Neuroscience, 760 Westwood Plaza, Suite A7-452 Los Angeles, CA, 90095, United States
| | - Aaron Wolfe Scheffler
- Department of Biostatistics, UCLA School of Public Health, Room 21-254C, CHS, Los Angeles, CA, 90095, United States
| | - Damla Senturk
- Department of Biostatistics, UCLA School of Public Health, Room 21-254C, CHS, Los Angeles, CA, 90095, United States
| | - Shafali Spurling Jeste
- Center for Autism Research and Treatment, University of California, Semel Institute for Neuroscience, 760 Westwood Plaza, Suite A7-452 Los Angeles, CA, 90095, United States
| |
Collapse
|
16
|
Sharma P, Powell KL, Wlodek ME, O'Brien TJ, Gilby KL. Delayed myelination and neurodevelopment in male seizure-prone versus seizure-resistant rats. Epilepsia 2018; 59:753-764. [PMID: 29377096 DOI: 10.1111/epi.14013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2018] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Aberrant myelination and developmental delay have been reported in epilepsy. However, it is unclear whether these are linked to intrinsic mechanisms that support a predisposition toward seizures and the development of epilepsy. Thus, we compared rates of myelination and neurodevelopment in male rats selectively bred for enhanced susceptibility to kindling epileptogenesis (FAST) with male rats bred for resistance (SLOW). METHODS Myelin-specific gene expression was compared in the brainstem, cerebellum, and cerebral hemisphere of FAST and SLOW rats on postnatal days (PNDs) 5, 11, 17, 23, and 90 to determine strain-specific myelination rates. Myelin protein levels were also compared at PNDs 5 and 23 in the brainstem. Relative rates of neurodevelopment were evaluated between PNDs 5 and 21 using physical growth landmarks and neuromotor tests including righting reflex, cliff avoidance, negative geotaxis, and locomotor activity. RESULTS Myelin-specific mRNA expression was significantly down-regulated in FAST rats on PNDs 5 and 11 in all 3 brain structures, indicating relatively delayed myelination. Likewise, corresponding protein levels were significantly lower in FAST brainstem on PND 5. Developmental delay was evident in the FAST strain such that only 9% of FAST pups, compared to 81% of SLOW, had open eyes by PND 13, locomotor activity was significantly reduced between PNDs 12 and 16, and neuromotor task acquisition was delayed between PNDs 5 and 10. SIGNIFICANCE Relative delays in myelination and neurodevelopment co-occurred in the seizure-prone FAST strain in the absence of seizures. These findings suggest these symptoms are not seizure-induced and may be mechanistically linked to an underlying pathophysiology supporting a predisposition toward developing epilepsy.
Collapse
Affiliation(s)
- Pragati Sharma
- Department of Medicine, University of Melbourne, Royal Melbourne Hospital, Parkville, Vic., Australia
| | - Kim L Powell
- Department of Medicine, University of Melbourne, Royal Melbourne Hospital, Parkville, Vic., Australia
| | - Mary E Wlodek
- Department of Physiology, University of Melbourne, Parkville, Vic., Australia
| | - Terence J O'Brien
- Department of Medicine, University of Melbourne, Royal Melbourne Hospital, Parkville, Vic., Australia
| | - Krista L Gilby
- Department of Medicine, University of Melbourne, Royal Melbourne Hospital, Parkville, Vic., Australia
| |
Collapse
|
17
|
Wang R, Wilkinson M, Kane T, Takahashi E. Convergence of Cortical, Thalamocortical, and Callosal Pathways during Human Fetal Development Revealed by Diffusion MRI Tractography. Front Neurosci 2017; 11:576. [PMID: 29163000 PMCID: PMC5671991 DOI: 10.3389/fnins.2017.00576] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 10/02/2017] [Indexed: 11/17/2022] Open
Abstract
There has been evidence that during brain development, emerging thalamocortical (TC) and corticothalamic (CT) pathways converge in some brain regions and follow each other's trajectories to their final destinations. Corpus callosal (CC) pathways also emerge at a similar developmental stage, and are known to converge with TC pathways in specific cortical regions in mature brains. Given the functional relationships between TC and CC pathways, anatomical convergence of the two pathways are likely important for their functional integration. However, it is unknown (1) where TC and CT subcortically converge in the human brain, and (2) where TC and CC converge in the cortex of the human brain, due to the limitations of non-invasive methods. The goals of this study were to describe the spatio-temporal relationships in the development of the TC/CT and CC pathways in the human brain, using high-angular resolution diffusion MR imaging (HARDI) tractography. Emerging cortical, TC and CC pathways were identified in postmortem fetal brains ranging from 17 gestational weeks (GW) to 30 GW, as well as in vivo 34-40 GW newborns. Some pathways from the thalami were found to be converged with pathways from the cerebral cortex as early as 17 GW. Such convergence was observed mainly in anterior and middle regions of the brain until 21 GW. At 22 GW and onwards, posterior pathways from the thalami also converged with cortical pathways. Many CC pathways reached the full length up to the cortical surface as early as 17 GW, while pathways linked to thalami (not only TC axons but also including pathways linked to thalamic neuronal migration) reached the cortical surface at and after 20 GW. These results suggest that CC pathways developed earlier than the TC pathways. The two pathways were widespread at early stages, but by 40 GW they condensed and formed groups of pathways that projected to specific regions of the cortex and overlapped in some brain regions. These results suggest that HARDI tractography has the potential to identify developing TC/CT and CC pathways with the timing and location of their convergence in fetal stages persisting in postnatal development.
Collapse
Affiliation(s)
- Rongpin Wang
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
- Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Molly Wilkinson
- Department of Behavioral Neuroscience, Northeastern University, Boston, MA, United States
| | - Tara Kane
- Department of Behavioral Neuroscience, Northeastern University, Boston, MA, United States
| | - Emi Takahashi
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
18
|
Environmental enrichment reduces brain damage in hydrocephalic immature rats. Childs Nerv Syst 2017; 33:921-931. [PMID: 28382436 DOI: 10.1007/s00381-017-3403-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 03/27/2017] [Indexed: 01/12/2023]
Abstract
PURPOSE We investigate the effects of environmental enrichment (EE) on morphological alterations in different brain structures of pup rats submitted to hydrocephalus condition. METHODS Hydrocephalus was induced in 7-day-old pup rats by injection of 20% kaolin into the cisterna magna. Ventricular dilatation and magnetization transfer to analyze myelin were assessed by magnetic resonance. Hydrocephalic and control rats exposed to EE (n = 10 per group) were housed in cages with a tunnel, ramp, and colored plastic balls that would emit sound when touched. The walls of the housing were decorated with colored adhesive tape. Moreover, tactile and auditory stimulation was performed daily throughout the experiment. Hydrocephalic and control rats not exposed to EE (n = 10 per group) were allocated singly in standard cages. All animals were weighed daily and exposed to open-field conditions every 2 days until the end of the experiment when they were sacrificed and the brains removed for histology and immunohistochemistry. Solochrome cyanine staining was performed to assess the thickness of the corpus callosum. The glial fibrillary acidic protein method was used to evaluate reactive astrocytes, and the Ki67 method to assess cellular proliferation in the subventricular zone. RESULTS The hydrocephalic animals exposed to EE showed better performance in Open Field tests (p < 0.05), while presenting lower weight gain. In addition, these animals showed better myelination as revealed by magnetization transfer (p < 0.05). Finally, the EE group showed a reduction in reactive astrocytes by means of glial fibrillary acidic protein immunostaining and preservation of the proliferation potential of progenitor cells. CONCLUSION The results suggest that EE can protect the developing brain against damaging effects caused by hydrocephalus.
Collapse
|
19
|
Khundrakpam BS, Lewis JD, Kostopoulos P, Carbonell F, Evans AC. Cortical Thickness Abnormalities in Autism Spectrum Disorders Through Late Childhood, Adolescence, and Adulthood: A Large-Scale MRI Study. Cereb Cortex 2017; 27:1721-1731. [PMID: 28334080 DOI: 10.1093/cercor/bhx038] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Indexed: 01/05/2023] Open
Abstract
Neuroimaging studies in autism spectrum disorders (ASDs) have provided inconsistent evidence of cortical abnormality. This is probably due to the small sample sizes used in most studies, and important differences in sample characteristics, particularly age, as well as to the heterogeneity of the disorder. To address these issues, we assessed abnormalities in ASD within the Autism Brain Imaging Data Exchange data set, which comprises data from approximately 1100 individuals (~6-55 years). A subset of these data that met stringent quality control and inclusion criteria (560 male subjects; 266 ASD; age = 6-35 years) were used to compute age-specific differences in cortical thickness in ASD and the relationship of any such differences to symptom severity of ASD. Our results show widespread increased cortical thickness in ASD, primarily left lateralized, from 6 years onwards, with differences diminishing during adulthood. The severity of symptoms related to social affect and communication correlated with these cortical abnormalities. These results are consistent with the conjecture that developmental patterns of cortical thickness abnormalities reflect delayed cortical maturation and highlight the dynamic nature of morphological abnormalities in ASD.
Collapse
Affiliation(s)
| | - John D Lewis
- Montreal Neurological Institute, McGill University, Montreal, QC, CanadaH3H2P1
| | | | - Felix Carbonell
- Montreal Neurological Institute, McGill University, Montreal, QC, CanadaH3H2P1
| | - Alan C Evans
- Montreal Neurological Institute, McGill University, Montreal, QC, CanadaH3H2P1
| |
Collapse
|
20
|
Magdalon J, Sánchez-Sánchez SM, Griesi-Oliveira K, Sertié AL. Dysfunctional mTORC1 Signaling: A Convergent Mechanism between Syndromic and Nonsyndromic Forms of Autism Spectrum Disorder? Int J Mol Sci 2017; 18:ijms18030659. [PMID: 28335463 PMCID: PMC5372671 DOI: 10.3390/ijms18030659] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/13/2017] [Accepted: 03/14/2017] [Indexed: 12/28/2022] Open
Abstract
Whereas autism spectrum disorder (ASD) exhibits striking heterogeneity in genetics and clinical presentation, dysfunction of mechanistic target of rapamycin complex 1 (mTORC1) signaling pathway has been identified as a molecular feature common to several well-characterized syndromes with high prevalence of ASD. Additionally, recent findings have also implicated mTORC1 signaling abnormalities in a subset of nonsyndromic ASD, suggesting that defective mTORC1 pathway may be a potential converging mechanism in ASD pathology across different etiologies. However, the mechanistic evidence for a causal link between aberrant mTORC1 pathway activity and ASD neurobehavioral features varies depending on the ASD form involved. In this review, we first discuss six monogenic ASD-related syndromes, including both classical and potentially novel mTORopathies, highlighting their contribution to our understanding of the neurobiological mechanisms underlying ASD, and then we discuss existing evidence suggesting that aberrant mTORC1 signaling may also play a role in nonsyndromic ASD.
Collapse
Affiliation(s)
- Juliana Magdalon
- Hospital Israelita Albert Einstein, Centro de Pesquisa Experimental, São Paulo 05652-900, Brazil.
| | - Sandra M Sánchez-Sánchez
- Hospital Israelita Albert Einstein, Centro de Pesquisa Experimental, São Paulo 05652-900, Brazil.
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo 05508-090, Brazil.
| | - Karina Griesi-Oliveira
- Hospital Israelita Albert Einstein, Centro de Pesquisa Experimental, São Paulo 05652-900, Brazil.
| | - Andréa L Sertié
- Hospital Israelita Albert Einstein, Centro de Pesquisa Experimental, São Paulo 05652-900, Brazil.
| |
Collapse
|
21
|
Tian Y, Wang L, Jia M, Lu T, Ruan Y, Wu Z, Wang L, Liu J, Zhang D. Association of oligodendrocytes differentiation regulator gene DUSP15 with autism. World J Biol Psychiatry 2017; 18:143-150. [PMID: 27223645 DOI: 10.1080/15622975.2016.1178395] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
OBJECTIVES Autism is a pervasive neurodevelopmental disorder with high heritability. Genetic factors play crucial roles in the aetiology of autism. Dual specificity phosphatase 15 (DUSP15) has been recognised as a key regulator gene for oligodendrocytes differentiation. A previous study detected one de novo missense variant (p.Thr107Met) with probable deleterious function in exon 6 of DUSP15 among patients with autism. Therefore, we sequenced this mutation in autistic children and performed an association analysis between DUSP15 polymorphisms and autism. METHODS We performed a case-control study between 255 children affected with autism and 427 healthy controls. Four tag-single nucleotide polymorphisms (SNPs) were selected. These SNPs and the previously reported mutation in exon 6 of DUSP15 were genotyped via Sanger sequencing. RESULTS Our results showed that rs3746599 was significantly associated with autism under allelic, additive and dominant models, respectively (χ2 = 9.699, P = 0.0018; χ2 = 16.224, P = 0.001; χ2 = 7.198, P = 0.007). The association remained significant after Bonferroni correction and permutation tests (n = 10,000). We did not detect the missense variant p.Thr107Met reported in previous studies. However, a de novo missense variant of DUSP15 (p.Ala56Thr) with a probable disease-causing effect was detected in one autistic child while absent in healthy controls. CONCLUSIONS Our findings initially suggest that DUSP15 might be a susceptibility gene for autism in Chinese Han population.
Collapse
Affiliation(s)
- Ye Tian
- a Institute of Mental Health, Peking University , Beijing , PR China.,b Peking University Sixth Hospital , Beijing , PR China.,c Key Laboratory for Mental Health , Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University) , Beijing , PR China
| | - Lifang Wang
- a Institute of Mental Health, Peking University , Beijing , PR China.,b Peking University Sixth Hospital , Beijing , PR China.,c Key Laboratory for Mental Health , Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University) , Beijing , PR China
| | - Meixiang Jia
- a Institute of Mental Health, Peking University , Beijing , PR China.,b Peking University Sixth Hospital , Beijing , PR China.,c Key Laboratory for Mental Health , Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University) , Beijing , PR China
| | - Tianlan Lu
- a Institute of Mental Health, Peking University , Beijing , PR China.,b Peking University Sixth Hospital , Beijing , PR China.,c Key Laboratory for Mental Health , Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University) , Beijing , PR China
| | - Yanyan Ruan
- a Institute of Mental Health, Peking University , Beijing , PR China.,b Peking University Sixth Hospital , Beijing , PR China.,c Key Laboratory for Mental Health , Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University) , Beijing , PR China
| | - Zhiliu Wu
- a Institute of Mental Health, Peking University , Beijing , PR China.,b Peking University Sixth Hospital , Beijing , PR China.,c Key Laboratory for Mental Health , Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University) , Beijing , PR China
| | - Linyan Wang
- a Institute of Mental Health, Peking University , Beijing , PR China.,b Peking University Sixth Hospital , Beijing , PR China.,c Key Laboratory for Mental Health , Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University) , Beijing , PR China
| | - Jing Liu
- a Institute of Mental Health, Peking University , Beijing , PR China.,b Peking University Sixth Hospital , Beijing , PR China.,c Key Laboratory for Mental Health , Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University) , Beijing , PR China
| | - Dai Zhang
- a Institute of Mental Health, Peking University , Beijing , PR China.,b Peking University Sixth Hospital , Beijing , PR China.,c Key Laboratory for Mental Health , Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University) , Beijing , PR China.,d PKU-IDG/McGovern Institute for Brain Research, Peking University , Beijing , PR China
| |
Collapse
|
22
|
Weickenmeier J, de Rooij R, Budday S, Steinmann P, Ovaert T, Kuhl E. Brain stiffness increases with myelin content. Acta Biomater 2016; 42:265-272. [PMID: 27475531 DOI: 10.1016/j.actbio.2016.07.040] [Citation(s) in RCA: 172] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 05/31/2016] [Accepted: 07/22/2016] [Indexed: 10/21/2022]
Abstract
UNLABELLED Brain stiffness plays an important role in neuronal development and disease, but reported stiffness values vary significantly for different species, for different brains, and even for different regions within the same brain. Despite extensive research throughout the past decade, the mechanistic origin of these stiffness variations remains elusive. Here we show that brain tissue stiffness is correlated to the underlying tissue microstructure and directly proportional to the local myelin content. In 116 indentation tests of six freshly harvested bovine brains, we found that the cerebral stiffnesses of 1.33±0.63kPa in white matter and 0.68±0.20kPa in gray matter were significantly different (p<0.01). Strikingly, while the inter-specimen variation was rather moderate, the minimum and maximum cerebral white matter stiffnesses of 0.59±0.19 kPa and 2.36±0.64kPa in each brain varied by a factor of four on average. To provide a mechanistic interpretation for this variation, we performed a histological characterization of the tested brain regions. We stained the samples with hematoxylin and eosin and luxol fast blue and quantified the local myelin content using image analysis. Interestingly, we found that the cerebral white matter stiffness increased with increasing myelin content, from 0.72kPa at a myelin content of 64-2.45kPa at a myelin content of 89%, with a Pearson correlation coefficient of ρ=0.91 (p<0.01). This direct correlation could have significant neurological implications. During development, our results could help explain why immature, incompletely myelinated brains are softer than mature, myelinated brains and more vulnerable to mechanical insult as evident, for example, in shaken baby syndrome. During demyelinating disease, our findings suggest to use stiffness alterations as clinical markers for demyelination to quantify the onset of disease progression, for example, in multiple sclerosis. Taken together, our study indicates that myelin might play a more important function than previously thought: It not only insulates signal propagation and improves electrical function of single axons, it also provides structural support and mechanical stiffness to the brain as a whole. STATEMENT OF SIGNIFICANCE Increasing evidence suggests that the mechanical environment of the brain plays an important role in neuronal development and disease. Reported stiffness values vary significantly, but the origin of these variations remains unknown. Here we show that stiffness of our brain is correlated to the underlying tissue microstructure and directly proportional to the local myelin content. Myelin has been discovered in 1854 as an insulating layer around nerve cells to improve electric signal propagation. Our study now shows that it also plays an important mechanical role: Using a combined mechanical characterization and histological characterization, we found that the white matter stiffness increases linearly with increasing myelin content, from 0.5kPa at a myelin content of 63-2.5kPa at 92%.
Collapse
|
23
|
Patak J, Hess JL, Zhang-James Y, Glatt SJ, Faraone SV. SLC9A9 Co-expression modules in autism-associated brain regions. Autism Res 2016; 10:414-429. [PMID: 27439572 DOI: 10.1002/aur.1670] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/27/2016] [Accepted: 06/13/2016] [Indexed: 12/16/2022]
Abstract
SLC9A9 is a sodium hydrogen exchanger present in the recycling endosome and highly expressed in the brain. It is implicated in neuropsychiatric disorders, including autism spectrum disorders (ASDs). Little research concerning its gene expression patterns and biological pathways has been conducted. We sought to investigate its possible biological roles in autism-associated brain regions throughout development. We conducted a weighted gene co-expression network analysis on RNA-seq data downloaded from Brainspan. We compared prenatal and postnatal gene expression networks for three ASD-associated brain regions known to have high SLC9A9 gene expression. We also performed an ASD-associated single nucleotide polymorphism enrichment analysis and a cell signature enrichment analysis. The modules showed differences in gene constituents (membership), gene number, and connectivity throughout time. SLC9A9 was highly associated with immune system functions, metabolism, apoptosis, endocytosis, and signaling cascades. Gene list comparison with co-immunoprecipitation data was significant for multiple modules. We found a disproportionately high autism risk signal among genes constituting the prenatal hippocampal module. The modules were enriched with astrocyte and oligodendrocyte markers. SLC9A9 is potentially involved in the pathophysiology of ASDs. Our investigation confirmed proposed functions for SLC9A9, such as endocytosis and immune regulation, while also revealing potential roles in mTOR signaling and cell survival.. By providing a concise molecular map and interactions, evidence of cell type and implicated brain regions we hope this will guide future research on SLC9A9. Autism Res 2017, 10: 414-429. © 2016 International Society for Autism Research, Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jameson Patak
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, New York
| | - Jonathan L Hess
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, New York
| | - Yanli Zhang-James
- Department of Psychiatry, Upstate Medical University, Syracuse, New York
| | - Stephen J Glatt
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, New York.,Department of Psychiatry, Upstate Medical University, Syracuse, New York
| | - Stephen V Faraone
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, New York.,Department of Psychiatry, Upstate Medical University, Syracuse, New York.,Department of Biomedicine, K.G. Jebsen Centre for Neuropsychiatric Disorders, University of Bergen, Bergen, Norway
| |
Collapse
|
24
|
Up-Regulation of Oligodendrocyte Lineage Markers in the Cerebellum of Autistic Patients: Evidence from Network Analysis of Gene Expression. Mol Neurobiol 2015; 53:4019-4025. [PMID: 26189831 DOI: 10.1007/s12035-015-9351-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 07/07/2015] [Indexed: 12/21/2022]
Abstract
Autism is a neurodevelopmental disorder manifested by impaired social interaction, deficits in communication skills, restricted interests, and repetitive behaviors. In neurodevelopmental, neurodegenerative, and psychiatric disorders, glial cells undergo morphological, biochemical, and functional rearrangements, which are critical for neuronal development, neurotransmission, and synaptic connectivity. Cerebellar function is not limited to motor coordination but also contributes to cognition and may be affected in autism. Oligodendrocytes and specifically oligodendroglial precursors are highly susceptible to oxidative stress and excitotoxic insult. In the present study, we searched for evidence for developmental oligodendropathy in the context of autism by performing a network analysis of gene expression of cerebellar tissue. We created an in silico network model (OLIGO) showing the landscape of interactions between oligodendrocyte markers and demonstrated that more than 50 % (16 out of 30) of the genes within this model displayed significant changes of expression (corrected p value <0.05) in the cerebellum of autistic patients. In particular, we found up-regulation of OLIG2-, MBP-, OLIG1-, and MAG-specific oligodendrocyte markers. We postulate that aberrant expression of oligodendrocyte-specific genes, potentially related to changes in oligodendrogenesis, may contribute to abnormal cerebellar development, impaired myelination, and anomalous synaptic connectivity in autism spectrum disorders (ASD).
Collapse
|
25
|
Wolff JJ, Gerig G, Lewis JD, Soda T, Styner MA, Vachet C, Botteron KN, Elison JT, Dager SR, Estes AM, Hazlett HC, Schultz RT, Zwaigenbaum L, Piven J. Altered corpus callosum morphology associated with autism over the first 2 years of life. Brain 2015; 138:2046-58. [PMID: 25937563 DOI: 10.1093/brain/awv118] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/06/2015] [Indexed: 11/13/2022] Open
Abstract
Numerous brain imaging studies indicate that the corpus callosum is smaller in older children and adults with autism spectrum disorder. However, there are no published studies examining the morphological development of this connective pathway in infants at-risk for the disorder. Magnetic resonance imaging data were collected from 270 infants at high familial risk for autism spectrum disorder and 108 low-risk controls at 6, 12 and 24 months of age, with 83% of infants contributing two or more data points. Fifty-seven children met criteria for ASD based on clinical-best estimate diagnosis at age 2 years. Corpora callosa were measured for area, length and thickness by automated segmentation. We found significantly increased corpus callosum area and thickness in children with autism spectrum disorder starting at 6 months of age. These differences were particularly robust in the anterior corpus callosum at the 6 and 12 month time points. Regression analysis indicated that radial diffusivity in this region, measured by diffusion tensor imaging, inversely predicted thickness. Measures of area and thickness in the first year of life were correlated with repetitive behaviours at age 2 years. In contrast to work from older children and adults, our findings suggest that the corpus callosum may be larger in infants who go on to develop autism spectrum disorder. This result was apparent with or without adjustment for total brain volume. Although we did not see a significant interaction between group and age, cross-sectional data indicated that area and thickness differences diminish by age 2 years. Regression data incorporating diffusion tensor imaging suggest that microstructural properties of callosal white matter, which includes myelination and axon composition, may explain group differences in morphology.
Collapse
Affiliation(s)
- Jason J Wolff
- 1 Department of Educational Psychology, University of Minnesota, Minneapolis, MN, USA
| | - Guido Gerig
- 2 Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, USA
| | - John D Lewis
- 3 Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Takahiro Soda
- 4 Health Sciences and Technology, Harvard Medical School and Massachusetts Institute of Technology, Boston, MA, USA 5 Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Martin A Styner
- 5 Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA 6 Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Clement Vachet
- 2 Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, USA
| | - Kelly N Botteron
- 7 Department of Psychiatry, Washington University at St. Louis, St. Louis, MO, USA
| | - Jed T Elison
- 8 Institute for Child Development, University of Minnesota, Minneapolis, MN, USA
| | - Stephen R Dager
- 9 Department of Radiology, University of Washington, Seattle, WA, USA
| | - Annette M Estes
- 10 Department of Speech and Hearing Science, University of Washington, Seattle, WA, USA
| | - Heather C Hazlett
- 5 Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA 6 Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Robert T Schultz
- 11 Centre for Autism Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lonnie Zwaigenbaum
- 12 Department of Paediatrics, University of Alberta, Edmonton AB, Canada
| | - Joseph Piven
- 5 Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA 6 Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | |
Collapse
|
26
|
Hutsler JJ, Casanova MF. Review: Cortical construction in autism spectrum disorder: columns, connectivity and the subplate. Neuropathol Appl Neurobiol 2015; 42:115-34. [PMID: 25630827 DOI: 10.1111/nan.12227] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 01/16/2015] [Indexed: 01/28/2023]
Abstract
The cerebral cortex undergoes protracted maturation during human development and exemplifies how biology and environment are inextricably intertwined in the construction of complex neural circuits. Autism spectrum disorders are characterized by a number of pathological changes arising from this developmental process. These include: (i) alterations to columnar structure that have significant implications for the organization of cortical circuits and connectivity; (ii) alterations to synaptic spines on individual cortical units that may underlie specific types of connectional changes; and (iii) alterations within the cortical subplate, a region that plays a role in proper cortical development and in regulating interregional communication in the mature brain. Although the cerebral cortex is not the only structure affected in the disorder, it is a fundamental contributor to the behaviours that characterize autism. These alterations to cortical circuitry likely underlie the behavioural phenotype in autism and contribute to the unique pattern of deficits and strengths that characterize cognitive functioning. Recent findings within the cortical subplate may indicate that alterations to cortical construction begin prenatally, before activity-dependent connections are established, and are in need of further study. A better understanding of cortical development in autism spectrum disorders will draw bridges between the microanatomical computational circuitry and the atypical behaviours that arise when that circuitry is modified. In addition, it will allow us to better exploit the constructional plasticity within the brain to design more targeted interventions that better manage atypical cortical construction and that can be applied very early in postnatal life.
Collapse
Affiliation(s)
- Jeffrey J Hutsler
- Department of Psychology, Program in Neuroscience, University of Nevada, Reno, USA
| | - Manuel F Casanova
- Department of Psychiatry and Behavioral Science, University of Louisville School of Medicine, Louisville, USA
| |
Collapse
|
27
|
Elevated GFAP Protein in Anterior Cingulate Cortical White Matter in Males With Autism Spectrum Disorder. Autism Res 2015; 8:649-57. [DOI: 10.1002/aur.1480] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 02/28/2015] [Indexed: 01/01/2023]
|
28
|
Berbel P, Navarro D, Román GC. An evo-devo approach to thyroid hormones in cerebral and cerebellar cortical development: etiological implications for autism. Front Endocrinol (Lausanne) 2014; 5:146. [PMID: 25250016 PMCID: PMC4158880 DOI: 10.3389/fendo.2014.00146] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Accepted: 08/25/2014] [Indexed: 12/11/2022] Open
Abstract
The morphological alterations of cortical lamination observed in mouse models of developmental hypothyroidism prompted the recognition that these experimental changes resembled the brain lesions of children with autism; this led to recent studies showing that maternal thyroid hormone deficiency increases fourfold the risk of autism spectrum disorders (ASD), offering for the first time the possibility of prevention of some forms of ASD. For ethical reasons, the role of thyroid hormones on brain development is currently studied using animal models, usually mice and rats. Although mammals have in common many basic developmental principles regulating brain development, as well as fundamental basic mechanisms that are controlled by similar metabolic pathway activated genes, there are also important differences. For instance, the rodent cerebral cortex is basically a primary cortex, whereas the primary sensory areas in humans account for a very small surface in the cerebral cortex when compared to the associative and frontal areas that are more extensive. Associative and frontal areas in humans are involved in many neurological disorders, including ASD, attention deficit-hyperactive disorder, and dyslexia, among others. Therefore, an evo-devo approach to neocortical evolution among species is fundamental to understand not only the role of thyroid hormones and environmental thyroid disruptors on evolution, development, and organization of the cerebral cortex in mammals but also their role in neurological diseases associated to thyroid dysfunction.
Collapse
Affiliation(s)
- Pere Berbel
- Departamento de Histología y Anatomía, Facultad de Medicina, Universidad Miguel Hernández, Alicante, Spain
| | - Daniela Navarro
- Departamento de Histología y Anatomía, Facultad de Medicina, Universidad Miguel Hernández, Alicante, Spain
| | - Gustavo C. Román
- Department of Neurology, Weill Cornell Medical College, Cornell University, New York, NY, USA
- Methodist Neurological Institute, Houston, TX, USA
| |
Collapse
|
29
|
Dysregulation of the IGF-I/PI3K/AKT/mTOR signaling pathway in autism spectrum disorders. Int J Dev Neurosci 2014; 35:35-41. [PMID: 24662006 DOI: 10.1016/j.ijdevneu.2014.03.006] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 03/13/2014] [Accepted: 03/13/2014] [Indexed: 11/20/2022] Open
Abstract
The IGF-I/PI3K/AKT/mTOR signaling pathway plays an important role in the regulation of cell growth, proliferation, differentiation, motility, survival, metabolism and protein synthesis. Insulin-like growth factor-I (IGF-I) is synthesized in the liver and fibroblasts, and its biological actions are mediated by the IGF-I receptor (IGF-IR). The binding of IGF-I to IGF-IR leads to the activation of phosphatidylinositol 3-kinase (PI3K). Activated PI3K stimulates the production of phosphatidylinositol (4,5)-bisphosphate [PI(4,5)P2] and phosphatidylinositol (3,4,5)-trisphosphate [PI(3,4,5)P3]. The PH domain of AKT (protein kinase B, PKB) (v-AKT murine thymoma viral oncogene homolog) binds to PI(4,5)P2 and PI(3,4,5)P3, followed by phosphorylation of the Thr308 and Ser473 regulatory sites. Tuberous sclerosis complex 1 (TSC1) and TSC2 are upstream regulators of mammalian target of rapamycin (mTOR) and downstream effectors of the PI3K/AKT signaling pathway. The activation of AKT suppresses the TSC1/TSC2 heterodimer, which is an upstream regulator of mTOR. Dysregulated IGF-I/PI3K/AKT/mTOR signaling has been shown to be associated with autism spectrum disorders (ASDs). In this review, we discuss the emerging evidence for a functional relationship between the IGF-I/PI3K/AKT/mTOR pathway and ASDs, as well as a possible role of this signaling pathway in the diagnosis and treatment of ASDs.
Collapse
|
30
|
Carson AM, Salowitz NMG, Scheidt RA, Dolan BK, Van Hecke AV. Electroencephalogram coherence in children with and without autism spectrum disorders: decreased interhemispheric connectivity in autism. Autism Res 2014; 7:334-43. [PMID: 24623657 DOI: 10.1002/aur.1367] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 01/24/2014] [Indexed: 11/11/2022]
Abstract
Electroencephalogram coherence was measured in children with autism spectrum disorders (ASD) and control children at baseline and while watching videos of a familiar and unfamiliar person reading a story. Coherence was measured between the left and right hemispheres of the frontal, parietal, and temporal-parietal lobes (interhemispheric) and between the frontal and parietal lobes in each hemisphere (intrahemispheric). A data-reduction technique was employed to identify the frequency (alpha) that yielded significant differences in video conditions. Children with ASD displayed reduced coherence at the alpha frequency between the left and right temporal-parietal lobes in all conditions and reduced coherence at the alpha frequency between left and right frontal lobes during baseline. No group differences in intrahemispheric coherence at the alpha frequency emerged at the chosen statistical threshold. Results suggest decreased interhemispheric connectivity in frontal and temporal-parietal regions in children with ASD compared to controls.
Collapse
Affiliation(s)
- Audrey M Carson
- Psychology Service, Baylor College of Medicine/Texas Children's Hospital, Houston, Texas, USA
| | | | | | | | | |
Collapse
|
31
|
Latchney SE, Masiulis I, Zaccaria KJ, Lagace DC, Powell CM, McCasland JS, Eisch AJ. Developmental and adult GAP-43 deficiency in mice dynamically alters hippocampal neurogenesis and mossy fiber volume. Dev Neurosci 2014; 36:44-63. [PMID: 24576816 DOI: 10.1159/000357840] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 12/09/2013] [Indexed: 12/29/2022] Open
Abstract
Growth-associated protein-43 (GAP-43) is a presynaptic protein that plays key roles in axonal growth and guidance and in modulating synapse formation. Previous work has demonstrated that mice lacking one allele of this gene (GAP-43+/- mice) exhibit hippocampal structural abnormalities, impaired spatial learning and stress-induced behavioral withdrawal and anxiety, behaviors that are dependent on proper hippocampal circuitry and function. Given the correlation between hippocampal function, synaptic connectivity and neurogenesis, we tested if behaviorally naïve GAP-43+/- mice had alterations in either neurogenesis or synaptic connectivity in the hippocampus during early postnatal development and young adulthood, and following behavior testing in older adults. To test our hypothesis, we examined hippocampal cell proliferation (Ki67), number of immature neuroblasts (doublecortin, DCX) and mossy fiber volume (synaptoporin) in behaviorally naïve postnatal day 9 (P9) and P26, and behaviorally experienced 5- to 7-month-old GAP-43+/- and +/+ littermate mice. P9 GAP-43+/- mice had fewer Ki67+ and DCX+ cells compared to +/+ mice, particularly in the posterior dentate gyrus, and smaller mossy fiber volume in the same region. In young adulthood, however, male GAP-43+/- mice had more Ki67+ and DCX+ cells and greater mossy fiber volume in the posterior dentate gyrus relative to male +/+ mice. These increases were not seen in females. In 5- to 7-month-old GAP-43+/- mice (whose behaviors were the focus of our prior publication), there was no global change in the number of proliferating or immature neurons relative to +/+ mice. However, more detailed analysis revealed fewer proliferative DCX+ cells in the anterior dentate gyrus of male GAP-43+/- mice compared to male +/+ mice. This reduction was not observed in females. These results suggest that young GAP-43+/- mice have decreased hippocampal neurogenesis and synaptic connectivity, but slightly older mice have greater hippocampal neurogenesis and synaptic connectivity. In conjunction with our previous study, these findings suggest that GAP-43 is dynamically involved in early postnatal and adult hippocampal neurogenesis and synaptic connectivity, possibly contributing to the GAP-43+/- behavioral phenotype.
Collapse
Affiliation(s)
- Sarah E Latchney
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Tex., USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Raznahan A, Wallace GL, Antezana L, Greenstein D, Lenroot R, Thurm A, Gozzi M, Spence S, Martin A, Swedo SE, Giedd JN. Compared to what? Early brain overgrowth in autism and the perils of population norms. Biol Psychiatry 2013; 74:563-75. [PMID: 23706681 PMCID: PMC4837958 DOI: 10.1016/j.biopsych.2013.03.022] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 02/27/2013] [Accepted: 03/13/2013] [Indexed: 12/14/2022]
Abstract
BACKGROUND Early brain overgrowth (EBO) in autism spectrum disorder (ASD) is among the best replicated biological associations in psychiatry. Most positive reports have compared head circumference (HC) in ASD (an excellent proxy for early brain size) with well-known reference norms. We sought to reappraise evidence for the EBO hypothesis given 1) the recent proliferation of longitudinal HC studies in ASD, and 2) emerging reports that several of the reference norms used to define EBO in ASD may be biased toward detecting HC overgrowth in contemporary samples of healthy children. METHODS Systematic review of all published HC studies in children with ASD. Comparison of 330 longitudinally gathered HC measures between birth and 18 months from male children with autism (n = 35) and typically developing control subjects (n = 22). RESULTS In systematic review, comparisons with locally recruited control subjects were significantly less likely to identify EBO in ASD than norm-based studies (p < .001). Through systematic review and analysis of new data, we replicate seminal reports of EBO in ASD relative to classical HC norms but show that this overgrowth relative to norms is mimicked by patterns of HC growth age in a large contemporary community-based sample of US children (n ~ 75,000). Controlling for known HC norm biases leaves inconsistent support for a subtle, later emerging and subgroup specific pattern of EBO in clinically ascertained ASD versus community control subjects. CONCLUSIONS The best-replicated aspects of EBO reflect generalizable HC norm biases rather than disease-specific biomarkers. The potential HC norm biases we detail are not specific to ASD research but apply throughout clinical and academic medicine.
Collapse
Affiliation(s)
- Armin Raznahan
- Child Psychiatry Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland.
| | | | - Ligia Antezana
- Laboratory of Brain and Cognition, NIMH, NIH, Bethesda, MD, USA
| | | | - Rhoshel Lenroot
- Department of Psychiatry, University of New South Wales, Sydney, Australia
| | - Audrey Thurm
- Pediatric Developmental Neurosciences Branch, NIMH, NIH, Bethesda, MD, USA
| | - Marta Gozzi
- Pediatric Developmental Neurosciences Branch, NIMH, NIH, Bethesda, MD, USA
| | - Sarah Spence
- Department of Neurology, Children’s Hospital Boston, Harvard Medical School, MA, USA
| | - Alex Martin
- Laboratory of Brain and Cognition, NIMH, NIH, Bethesda, MD, USA
| | - Susan E Swedo
- Pediatric Developmental Neurosciences Branch, NIMH, NIH, Bethesda, MD, USA
| | - Jay N Giedd
- Child Psychiatry Branch, NIMH, NIH, Bethesda, MD, USA
| |
Collapse
|
33
|
Splenium of corpus callosum: patterns of interhemispheric interaction in children and adults. Neural Plast 2013; 2013:639430. [PMID: 23577273 PMCID: PMC3610378 DOI: 10.1155/2013/639430] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 02/08/2013] [Accepted: 02/09/2013] [Indexed: 01/09/2023] Open
Abstract
The splenium of the corpus callosum connects the posterior cortices with fibers varying in size from thin late-myelinating axons in the anterior part, predominantly connecting parietal and temporal areas, to thick early-myelinating fibers in the posterior part, linking primary and secondary visual areas. In the adult human brain, the function of the splenium in a given area is defined by the specialization of the area and implemented via excitation and/or suppression of the contralateral homotopic and heterotopic areas at the same or different level of visual hierarchy. These mechanisms are facilitated by interhemispheric synchronization of oscillatory activity, also supported by the splenium. In postnatal ontogenesis, structural MRI reveals a protracted formation of the splenium during the first two decades of human life. In doing so, the slow myelination of the splenium correlates with the formation of interhemispheric excitatory influences in the extrastriate areas and the EEG synchronization, while the gradual increase of inhibitory effects in the striate cortex is linked to the local inhibitory circuitry. Reshaping interactions between interhemispherically distributed networks under various perceptual contexts allows sparsification of responses to superfluous information from the visual environment, leading to a reduction of metabolic and structural redundancy in a child's brain.
Collapse
|
34
|
Lombardo MV, Chakrabarti B, Lai MC. Self-referential and social cognition in a case of autism and agenesis of the corpus callosum. Mol Autism 2012; 3:14. [PMID: 23171505 PMCID: PMC3522057 DOI: 10.1186/2040-2392-3-14] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 10/30/2012] [Indexed: 01/23/2023] Open
Abstract
UNLABELLED BACKGROUND While models of autism spectrum conditions (ASC) are emerging at the genetic level of analysis, clear models at higher levels of analysis, such as neuroanatomy, are lacking. Here we examine agenesis of the corpus callosum (AgCC) as a model at the level of neuroanatomy that may be relevant for understanding self-referential and social-cognitive difficulties in ASC. METHODS We examined performance on a wide array of tests in self-referential and social-cognitive domains in a patient with both AgCC and a diagnosis of ASC. Tests included a depth-of-processing memory paradigm with self-referential and social-cognitive manipulations, self-report measures of self-consciousness, alexithymia, and empathy, as well as performance measures of first-person pronoun usage and mentalizing ability. The performance of the AgCC patient was compared to a group of individuals with ASC but without AgCC and with neurotypical controls. These comparison groups come from a prior study where group differences were apparent across many measures. We used bootstrapping to assess whether the AgCC patient exhibited scores that were within or outside the 95% bias-corrected and accelerated bootstrap confidence intervals observed in both comparison groups. RESULTS Within the depth-of-processing memory paradigm, the AgCC patient showed decreased memory sensitivity that was more extreme than both comparison groups across all conditions. The patient's most pronounced difficulty on this task emerged in the social-cognitive domain related to information-processing about other people. The patient was similar to the ASC group in benefiting less from self-referential processing compared to the control group. Across a variety of other self-referential (i.e. alexithymia, private self-consciousness) and social-cognitive measures (i.e. self-reported imaginative and perspective-taking subscales of empathy, mentalizing), the AgCC patient also showed more extreme scores than those observed for both of the comparison groups. However, the AgCC patient scored within the range observed in the comparison groups on measures of first-person pronoun usage and self-reported affective empathy subscales. CONCLUSIONS We conclude that AgCC co-occurring with a diagnosis of ASC may be a relevant model at the level of neuroanatomy for understanding mechanisms involved in self-referential and high-level social-cognitive difficulties in ASC.
Collapse
Affiliation(s)
- Michael V Lombardo
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Douglas House, 18B Trumpington Road, Cambridge CB2 8AH, UK
| | - Bhismadev Chakrabarti
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Douglas House, 18B Trumpington Road, Cambridge CB2 8AH, UK
- Centre for Integrative Neuroscience and Neurodynamics, School of Psychology and Clinical Language Sciences, University of Reading, Whiteknights RG6 6AL, UK
| | - Meng-Chuan Lai
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Douglas House, 18B Trumpington Road, Cambridge CB2 8AH, UK
| |
Collapse
|