1
|
Verpeut JL, Oostland M. The significance of cerebellar contributions in early-life through aging. Front Comput Neurosci 2024; 18:1449364. [PMID: 39258107 PMCID: PMC11384999 DOI: 10.3389/fncom.2024.1449364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/12/2024] [Indexed: 09/12/2024] Open
Affiliation(s)
- Jessica L Verpeut
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | - Marlies Oostland
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
2
|
Chen Y, Bury LA, Chen F, Aldinger KA, Miranda HC, Wynshaw-Boris A. Generation of advanced cerebellar organoids for neurogenesis and neuronal network development. Hum Mol Genet 2023; 32:2832-2841. [PMID: 37387247 PMCID: PMC10481094 DOI: 10.1093/hmg/ddad110] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 06/26/2023] [Indexed: 07/01/2023] Open
Abstract
Neurons within the cerebellum form temporal-spatial connections through the cerebellum, and the entire brain. Organoid models provide an opportunity to model the early differentiation of the developing human cerebellum, which is difficult to study in vivo, and affords the opportunity to study neurodegenerative and neurodevelopmental diseases of the cerebellum. Previous cerebellar organoid models focused on early neuron generation and single cell activity. Here, we modify previous protocols to generate more mature cerebellar organoids that allow for the establishment of several classes of mature neurons during cerebellar differentiation and development, including the establishment of neural networks during whole-organoid maturation. This will provide a means to study the generation of several more mature cerebellar cell types, including Purkinje cells, granule cells and interneurons expression as well as neuronal communication for biomedical, clinical and pharmaceutical applications.
Collapse
Affiliation(s)
- Ya Chen
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Luke A Bury
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Fu Chen
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Kimberly A Aldinger
- Department of Pediatrics and Neurology, Center for Integrative Brain Research, Seattle Children’s Research Institute, University of Washington, Seattle, WA 98101, USA
| | - Helen C Miranda
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Anthony Wynshaw-Boris
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
3
|
Ocasio JK. Dissociation of Cerebellar Granule Neuron Progenitors for Culture, FACS, Transcriptomics, and Molecular Biology. Methods Mol Biol 2023; 2583:3-7. [PMID: 36418720 DOI: 10.1007/978-1-0716-2752-5_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Brain growth reflects the proliferation dynamics of neural progenitors, and understanding brain growth requires molecular, genetic, and functional studies of these specific cells. Cerebellar granule neuron progenitors (CGNPs) proliferate in the early postnatal period in both mice and humans, to generate the largest population of neurons in the central nervous system. CGNPs present a large, spatially segregated source of neural progenitors with a consistent, well-characterized temporal pattern of proliferation and differentiation that facilitates analysis. Dissociating of CGNPs with the methods below will generate a suspension of primary neural progenitors harvested from the postnatal brain that may be used for diverse experimental analyses including cell culture, protein extraction, flow cytometry, metabolomic analysis, and transcriptomic analysis with single-cell resolution (scRNA-seq).
Collapse
|
4
|
Iskusnykh IY, Chizhikov VV. Cerebellar development after preterm birth. Front Cell Dev Biol 2022; 10:1068288. [PMID: 36523506 PMCID: PMC9744950 DOI: 10.3389/fcell.2022.1068288] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/09/2022] [Indexed: 11/30/2022] Open
Abstract
Preterm birth and its complications and the associated adverse factors, including brain hemorrhage, inflammation, and the side effects of medical treatments, are the leading causes of neurodevelopmental disability. Growing evidence suggests that preterm birth affects the cerebellum, which is the brain region involved in motor coordination, cognition, learning, memory, and social communication. The cerebellum is particularly vulnerable to the adverse effects of preterm birth because key cerebellar developmental processes, including the proliferation of neural progenitors, and differentiation and migration of neurons, occur in the third trimester of a human pregnancy. This review discusses the negative impacts of preterm birth and its associated factors on cerebellar development, focusing on the cellular and molecular mechanisms that mediate cerebellar pathology. A better understanding of the cerebellar developmental mechanisms affected by preterm birth is necessary for developing novel treatment and neuroprotective strategies to ameliorate the cognitive, behavioral, and motor deficits experienced by preterm subjects.
Collapse
|
5
|
Raslan IR, Barsottini OG, Pedroso JL. A Proposed Clinical Classification and a Diagnostic Approach for Congenital Ataxias. Neurol Clin Pract 2021; 11:e328-e336. [PMID: 34484907 DOI: 10.1212/cpj.0000000000000966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/03/2020] [Indexed: 01/12/2023]
Abstract
Purpose of Review This review proposes a clinical classification for congenital ataxias based on clinical features, neuroimaging, and course of the disease. Recent Findings Congenital ataxias are an unusual group of neurologic disorders, with heterogeneous clinical and genetic presentation. Typical clinical features of congenital ataxias include variable degrees of motor developmental delay, very early onset cerebellar ataxia, cognitive impairment, and hypotonia, frequently mistakenly diagnosed as cerebral palsy. Congenital ataxias are usually nonprogressive. Neuroimaging plays an important role in the characterization of congenital ataxias. Despite the development of genetics with exome sequencing, several congenital ataxias remain undetermined, and medical literature on this topic is scarce. Summary A didactic classification based on the clinical and neuroimaging features for congenital ataxias include the following 4 main groups: cerebellar malformation, syndromic congenital ataxias, congenital cerebellar hypoplasia, and pontocerebellar hypoplasia. A diagnostic approach for congenital ataxias is proposed, and its differential diagnosis is also discussed.
Collapse
Affiliation(s)
- Ivana Rocha Raslan
- Ataxia Unit, Department of Neurology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Orlando G Barsottini
- Ataxia Unit, Department of Neurology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - José Luiz Pedroso
- Ataxia Unit, Department of Neurology, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
6
|
Abstract
Cerebellar hypoplasia (CH) refers to a cerebellum of reduced volume with preserved shape. CH is associated with a broad heterogeneity in neuroradiologic features, etiologies, clinical characteristics, and neurodevelopmental outcomes, challenging physicians evaluating children with CH. Traditionally, neuroimaging has been a key tool to categorize CH based on the pattern of cerebellar involvement (e.g., hypoplasia of cerebellar vermis only vs. hypoplasia of both the vermis and cerebellar hemispheres) and the presence of associated brainstem and cerebral anomalies. With the advances in genetic technologies of the recent decade, many novel CH genes have been identified, and consequently, a constant updating of the literature and revision of the classification of cerebellar malformations are needed. Here, we review the current literature on CH. We propose a systematic approach to recognize specific neuroimaging patterns associated with CH, based on whether the CH is isolated or associated with posterior cerebrospinal fluid anomalies, specific brainstem or cerebellar malformations, brainstem hypoplasia with or without cortical migration anomalies, or dysplasia. The CH radiologic pattern and clinical assessment will allow the clinician to guide his investigations and genetic testing, give a more precise diagnosis, screen for associated comorbidities, and improve prognostication of associated neurodevelopmental outcomes.
Collapse
|
7
|
Abstract
Astrocytes, initially described as merely support cells, are now known as a heterogeneous population of cells actively involved in a variety of biological functions such as: neuronal migration and differentiation; regulation of cerebral blood flow; metabolic control of extracellular potassium concentration; and modulation of synapse formation and elimination; among others. Cerebellar glial cells have been shown to play a significant role in proliferation, differentiation, migration, and synaptogenesis. However, less evidence is available about the role of neuron-astrocyte interactions during cerebellar development and their impact on diseases of the cerebellum. In this review, we will focus on the mechanisms underlying cellular interactions, specifically neuron-astrocyte interactions, during cerebellar development, function, and disease. We will discuss how cerebellar glia, astrocytes, and Bergmann glia play a fundamental role in several steps of cerebellar development, such as granule cell migration, axonal growth, neuronal differentiation, and synapse formation, and in diseases associated with the cerebellum. We will focus on how astrocytes and thyroid hormones impact cerebellar development. Furthermore, we will provide evidence of how growth factors secreted by glial cells, such as epidermal growth factor and transforming growth factors, control cerebellar organogenesis. Finally, we will argue that glia are a key mediator of cerebellar development and that identification of molecules and pathways involved in neuron-glia interactions may contribute to a better understanding of cerebellar development and associated disorders.
Collapse
|
8
|
Jandeaux C, Kuchcinski G, Ternynck C, Riquet A, Leclerc X, Pruvo JP, Soto-Ares G. Biometry of the Cerebellar Vermis and Brain Stem in Children: MR Imaging Reference Data from Measurements in 718 Children. AJNR Am J Neuroradiol 2019; 40:1835-1841. [PMID: 31624120 DOI: 10.3174/ajnr.a6257] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/21/2019] [Indexed: 01/04/2023]
Abstract
BACKGROUND AND PURPOSE Objective and quantitative data to define cerebellar vermis and/or brain stem hypoplasia in children are lacking. Our aim was to provide MR imaging biometric references for the cerebellar vermis and brain stem from a large cohort of children with normal cerebellums. MATERIALS AND METHODS The MR imaging data were retrospectively selected from our hospital data base from January 1, 2014, to December 31, 2017. All MR imaging examinations of children between 1 day and 15 years of age, including midline sagittal sections, were included. Children with a clinical history or MR imaging abnormalities that may affect the posterior fossa were excluded. We manually measured four 2D parameters: vermian height, anterior-posterior diameter of the vermis, anterior-posterior diameter of the midbrain-pons junction, and anterior-posterior midpons diameter. The inter- and intraobserver agreement was evaluated. RESULTS Seven hundred eighteen children were included (372 boys and 346 girls), from 1 day to 15 years of age. Normal values (third to 97th percentiles) were provided for each parameter. The vermis parameters showed a rapid growth phase during the first year, a slower growth until the fifth year, and finally a near-plateau phase. The brain stem parameters showed more progressive growth. The intra- and interobserver agreement was excellent for all parameters. CONCLUSIONS We provide reference biometric data of the vermis and the brain stem using simple and reproducible measurements that are easy to use in daily practice. The relevance of these 2D measurements should be further validated in diseases associated with cerebellar abnormalities.
Collapse
Affiliation(s)
- C Jandeaux
- From the Departments of Neuroradiology (C.J., G.K., X.L., J.-P.P., G.S.-A.)
| | - G Kuchcinski
- From the Departments of Neuroradiology (C.J., G.K., X.L., J.-P.P., G.S.-A.)
| | | | - A Riquet
- Neuropediatrics (A.R.), Centre Hospitalier Universitaire Lille, Lille, France
| | - X Leclerc
- From the Departments of Neuroradiology (C.J., G.K., X.L., J.-P.P., G.S.-A.)
| | - J-P Pruvo
- From the Departments of Neuroradiology (C.J., G.K., X.L., J.-P.P., G.S.-A.)
| | - G Soto-Ares
- From the Departments of Neuroradiology (C.J., G.K., X.L., J.-P.P., G.S.-A.)
| |
Collapse
|
9
|
Ocasio JK, Bates RDP, Rapp CD, Gershon TR. GSK-3 modulates SHH-driven proliferation in postnatal cerebellar neurogenesis and medulloblastoma. Development 2019; 146:dev.177550. [PMID: 31540917 DOI: 10.1242/dev.177550] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 09/04/2019] [Indexed: 12/28/2022]
Abstract
Cerebellar development requires regulated proliferation of cerebellar granule neuron progenitors (CGNPs). Inadequate CGNP proliferation causes cerebellar hypoplasia whereas excessive CGNP proliferation can cause medulloblastoma, the most common malignant pediatric brain tumor. Although sonic hedgehog (SHH) signaling is known to activate CGNP proliferation, the mechanisms downregulating proliferation are less defined. We investigated CGNP regulation by GSK-3, which downregulates proliferation in the forebrain, gut and breast by suppressing mitogenic WNT signaling in mouse. In striking contrast to these systems, we found that co-deleting Gsk3a and Gsk3b blocked CGNP proliferation, causing severe cerebellar hypoplasia. The GSK-3 inhibitor CHIR-98014 similarly downregulated SHH-driven proliferation. Transcriptomic analysis showed activated WNT signaling and upregulated Cdkn1a in Gsk3a/b -deleted CGNPs. Ctnnb co-deletion increased CGNP proliferation and rescued cerebellar hypoproliferation in Gsk3a/b mutants, demonstrating physiological control of CGNPs by GSK-3, mediated through WNT. SHH-driven medulloblastomas similarly required GSK-3, as co-deleting Gsk3a/b blocked tumor growth in medulloblastoma-prone SmoM2 mice. These data show that a GSK-3/WNT axis modulates the developmental proliferation of CGNPs and the pathological growth of SHH-driven medulloblastoma. The requirement for GSK-3 in SHH-driven proliferation suggests that GSK-3 may be targeted for SHH-driven medulloblastoma therapy.
Collapse
Affiliation(s)
- Jennifer K Ocasio
- UNC Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA .,Department of Neurology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Rolf Dale P Bates
- Department of Neurology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Carolyn D Rapp
- Department of Neurology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Timothy R Gershon
- UNC Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA .,Department of Neurology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
10
|
Mazzonetto PC, Ariza CB, Ocanha SG, de Souza TA, Ko GM, Menck CFM, Massironi SMG, Porcionatto MA. Mutation in NADPH oxidase 3 (NOX3) impairs SHH signaling and increases cerebellar neural stem/progenitor cell proliferation. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1502-1515. [PMID: 30853403 DOI: 10.1016/j.bbadis.2019.02.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 02/25/2019] [Accepted: 02/27/2019] [Indexed: 01/03/2023]
Abstract
Abnormalities in cerebellar structure and function may cause ataxia, a neurological dysfunction of motor coordination. In the course of the present study, we characterized a mutant mouse lineage with an ataxia-like phenotype. We localized the mutation on chromosome 17 and mapped it to position 1534 of the Nox3 gene, resulting in p.Asn64Tyr change. The primary defect observed in Nox3eqlb mice was increased proliferation of cerebellar granule cell precursors (GCPs). cDNA microarray comparing Nox3eqlb and BALB/c neonatal cerebellum revealed changes in the expression of genes involved in the control of cell proliferation. Nox3eqlb GCPs and NSC produce higher amounts of reactive oxygen species (ROS) and upregulate the expression of SHH target genes, such as Gli1-3 and Ccnd1 (CyclinD1). We hypothesize that this new mutation is responsible for an increase in proliferation via stimulation of the SHH pathway. We suggest this mutant mouse lineage as a new model to investigate the role of ROS in neuronal precursor cell proliferation.
Collapse
Affiliation(s)
- P C Mazzonetto
- Department of Biochemistry, Laboratory of Neurobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Brazil
| | - C B Ariza
- Department of Biochemistry, Laboratory of Neurobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Brazil; Department of General Pathology, Center of Biological Sciences, Universidade Estadual de Londrina (UEL), Brazil
| | - S G Ocanha
- Department of Biochemistry, Laboratory of Neurobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Brazil
| | - T A de Souza
- Department of Microbiology, Institute of Biomedical Sciences, Universidade de São Paulo (USP), Brazil
| | - G M Ko
- Department of Biochemistry, Laboratory of Neurobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Brazil
| | - C F M Menck
- Department of Microbiology, Institute of Biomedical Sciences, Universidade de São Paulo (USP), Brazil
| | - S M G Massironi
- Department of Immunology, Institute of Biomedical Sciences, Universidade de São Paulo (USP), Brazil
| | - M A Porcionatto
- Department of Biochemistry, Laboratory of Neurobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Brazil.
| |
Collapse
|
11
|
Shiohama T, Levman J, Takahashi E. Surface- and voxel-based brain morphologic study in Rett and Rett-like syndrome with MECP2 mutation. Int J Dev Neurosci 2019; 73:83-88. [PMID: 30690146 DOI: 10.1016/j.ijdevneu.2019.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 12/20/2018] [Accepted: 01/23/2019] [Indexed: 12/21/2022] Open
Abstract
Rett syndrome (RTT) is a rare congenital disorder which in most cases (95%) is caused by methyl-CpG binding protein 2 (MECP2) mutations. RTT is characterized by regression in global development, epilepsy, autistic features, acquired microcephaly, habitual hand clapping, loss of purposeful hand skills, and autonomic dysfunctions. Although the literature has demonstrated decreased volumes of the cerebrum, cerebellum, and the caudate nucleus in RTT patients, surface-based brain morphology including cortical thickness and cortical gyrification analyses are lacking in RTT. We present quantitative surface- and voxel-based morphological measurements in young children with RTT and Rett-like syndrome (RTT-l) with MECP2 mutations. The 8 structural T1-weighted MR images were obtained from 7 female patients with MECP2 mutations (3 classic RTT, 2 variant RTT, and 2 RTT-l) (mean age 5.2 [standard deviation 3.3] years old). Our analyses demonstrated decreased total volumes of the cerebellum in RTT/RTT-l compared to gender- and age-matched controls (t (22)=-2.93, p = .008, Cohen's d = 1.27). In contrast, global cerebral cortical surface areas, global/regional cortical thicknesses, the degree of global gyrification, and global/regional gray and white matter volumes were not statistically significantly different between the two groups. Our findings, as well as literature findings, suggest that early brain abnormalities associated with RTT/RTT-l (with MECP2 mutations) can be detected as regionally decreased cerebellar volumes. Decreased cerebellar volume may be helpful for understanding the etiology of RTT/RTT-l.
Collapse
Affiliation(s)
- Tadashi Shiohama
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, USA; Department of Pediatrics, Chiba University Hospital, Inohana 1-8-1, Chiba-shi, Chiba, 2608670, Japan.
| | - Jacob Levman
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, USA; Department of Mathematics, Statistics and Computer Science, St. Francis Xavier University, 2323 Notre Dame Ave, Antigonish, Nova Scotia, B2G 2W5, Canada
| | - Emi Takahashi
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, USA
| |
Collapse
|
12
|
Cerebellar Ataxia in Children: A Clinical and MRI Approach to the Differential Diagnosis. Top Magn Reson Imaging 2018; 27:275-302. [PMID: 30086112 DOI: 10.1097/rmr.0000000000000175] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
: The cerebellum has long been recognized as a fundamental structure in motor coordination. Structural cerebellar abnormalities and diseases involving the cerebellum are relatively common in children. The not always specific clinical presentation of ataxia, incoordination, and balance impairment can often be a challenge to attain a precise diagnosis. Continuous advances in genetic research and moreover the constant development in neuroimaging modalities, particularly in the field of magnetic resonance imaging, have promoted a better understanding of cerebellar diseases and led to several modifications in their classification in recent years. Thorough clinical and neuroimaging investigation is recommended for proper diagnosis. This review outlines an update of causes of cerebellar disorders that present clinically with ataxia in the pediatric population. These conditions were classified in 2 major groups, namely genetic malformations and acquired or disruptive disorders recognizable by neuroimaging and subsequently according to their features during the prenatal and postnatal periods.
Collapse
|
13
|
Iskusnykh IY, Buddington RK, Chizhikov VV. Preterm birth disrupts cerebellar development by affecting granule cell proliferation program and Bergmann glia. Exp Neurol 2018; 306:209-221. [PMID: 29772246 PMCID: PMC6291230 DOI: 10.1016/j.expneurol.2018.05.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 05/09/2018] [Accepted: 05/12/2018] [Indexed: 01/17/2023]
Abstract
Preterm birth is a leading cause of long-term motor and cognitive deficits. Clinical studies suggest that some of these deficits result from disruption of cerebellar development, but the mechanisms that mediate cerebellar abnormalities in preterm infants are largely unknown. Furthermore, it remains unclear whether preterm birth and precocious exposure to the ex-utero environment directly disrupt cerebellar development or indirectly by increasing the probability of cerebellar injury, including that resulting from clinical interventions and protocols associated with the care of preterm infants. In this study, we analyzed the cerebellum of preterm pigs delivered via c-section at 91% term and raised for 10 days, until term-equivalent age. The pigs did not receive any treatments known or suspected to affect cerebellar development and had no evidence of brain damage. Term pigs sacrificed at birth were used as controls. Immunohistochemical analysis revealed that preterm birth did not affect either size or numbers of Purkinje cells or molecular layer interneurons at term-equivalent age. The number of granule cell precursors and Bergmann glial fibers, however, were reduced in preterm pigs. Preterm pigs had reduced proliferation but not differentiation of granule cells. qRT-PCR analysis of laser capture microdissected external granule cell layer showed that preterm pigs had a reduced expression of Ccnd1 (Cyclin D1), Ccnb1 (Cyclin B1), granule cell master regulatory transcription factor Atoh1, and signaling molecule Jag1. In vitro rescue experiments identified Jag1 as a central granule cell gene affected by preterm birth. Thus, preterm birth and precocious exposure to the ex-utero environment disrupt cerebellum by modulating expression of key cerebellar developmental genes, predominantly affecting development of granule precursors and Bergmann glia.
Collapse
Affiliation(s)
- Igor Y Iskusnykh
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | | | - Victor V Chizhikov
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
14
|
Hadders-Algra M. Early human brain development: Starring the subplate. Neurosci Biobehav Rev 2018; 92:276-290. [PMID: 29935204 DOI: 10.1016/j.neubiorev.2018.06.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 06/15/2018] [Accepted: 06/19/2018] [Indexed: 12/16/2022]
Abstract
This review summarizes early human brain development on the basis of neuroanatomical data and functional connectomics. It indicates that the most significant changes in the brain occur during the second half of gestation and the first three months post-term, in particular in the cortical subplate and cerebellum. As the transient subplate pairs a high rate of intricate developmental changes and interactions with clear functional activity, two phases of development are distinguished: a) the transient cortical subplate phase, ending at 3 months post-term when the permanent circuitries in the primary motor, somatosensory and visual cortices have replaced the subplate; and subsequently, b) the phase in which the permanent circuitries dominate. In the association areas the subplate dissolves in the remainder of the first postnatal year. During both phases developmental changes are paralleled by continuous reconfigurations in network activity. The reviewed literature also suggests that disruption of subplate development may play a pivotal role in developmental disorders, such as cerebral palsy, autism spectrum disorders, attention deficit hyperactivity disorder and schizophrenia.
Collapse
Affiliation(s)
- Mijna Hadders-Algra
- University of Groningen, University Medical Center Groningen, Dept. Pediatrics - Section Developmental Neurology, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| |
Collapse
|
15
|
Adle-Biassette H, Golden JA, Harding B. Developmental and perinatal brain diseases. HANDBOOK OF CLINICAL NEUROLOGY 2018; 145:51-78. [PMID: 28987191 DOI: 10.1016/b978-0-12-802395-2.00006-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
This chapter briefly describes the normal development of the nervous system, the neuropathology and pathophysiology of acquired and secondary disorders affecting the embryo, fetus, and child. They include CNS manifestations of chromosomal change; forebrain patterning defects; disorders of the brain size; cell migration and specification disorders; cerebellum, hindbrain and spinal patterning defects; hydrocephalus; secondary malformations and destructive pathologies; vascular malformations; arachnoid cysts and infectious diseases. The distinction between malformations and disruptions is important for pathogenesis and genetic counseling.
Collapse
Affiliation(s)
- Homa Adle-Biassette
- Department of Pathology, Lariboisière Hospital, APHP and Paris Diderot University, Sorbonne Paris Cité, Paris, France.
| | - Jeffery A Golden
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Brian Harding
- Department of Pathology/Neuropathology, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| |
Collapse
|
16
|
Driver AM, Shumrick C, Stottmann RW. Ttc21b Is Required in Bergmann Glia for Proper Granule Cell Radial Migration. J Dev Biol 2017; 5:E18. [PMID: 29615573 PMCID: PMC5831799 DOI: 10.3390/jdb5040018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 11/30/2017] [Accepted: 12/11/2017] [Indexed: 12/11/2022] Open
Abstract
Proper cerebellar development is dependent on tightly regulated proliferation, migration, and differentiation events. Disruptions in any of these leads to a range of cerebellar phenotypes from ataxia to childhood tumors. Animal models have shown that proper regulation of sonic hedgehog (Shh) signaling is crucial for normal cerebellar architecture, and increased signaling leads to cerebellar tumor formation. Primary cilia are known to be required for the proper regulation of multiple developmental signaling pathways, including Shh. Tetratricopeptide Repeat Domain 21B (Ttc21b) is required for proper primary cilia form and function, and is primarily thought to restrict Shh signaling. Here we investigated a role for Ttc21b in cerebellar development. Surprisingly, Ttc21b ablation in Bergmann glia resulted in the accumulation of ectopic granule cells in the lower/posterior lobes of the cerebellum and a reduction in Shh signaling. Ttc21b ablation in just Purkinje cells resulted in a similar phenotype seen in fewer cells, but across the entire extent of the cerebellum. These results suggest that Ttc21b expression is required for Bergmann glia structure and signaling in the developing cerebellum, and in some contexts, augments rather than attenuates Shh signaling.
Collapse
Affiliation(s)
- Ashley M Driver
- Division of Human Genetics, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Christopher Shumrick
- Division of Human Genetics, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Rolf W Stottmann
- Division of Human Genetics, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
17
|
Hagan N, Guarente J, Ellisor D, Zervas M. The Temporal Contribution of the Gbx2 Lineage to Cerebellar Neurons. Front Neuroanat 2017; 11:50. [PMID: 28785208 PMCID: PMC5519623 DOI: 10.3389/fnana.2017.00050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 06/16/2017] [Indexed: 11/13/2022] Open
Abstract
The cerebellum (Cb) is an exquisite structure that controls elaborate motor behaviors and is essential for sensory-motor learning. During development, the Cb is derived from rhombomere 1 (r1). Within this embryonic compartment, precursors in r1 are patterned by signaling cues originating from the isthmus organizer (IsO) and subsequently undergo complex morphogenic movements to establish their final position in the mature Cb. The transcription factor Gbx2 is expressed in the developing Cb and is intimately involved in organizing and patterning the Cb. Nevertheless, how precursors expressing Gbx2 at specific embryonic time points contribute to distinct cell types in the adult Cb is unresolved. In this study, we used Genetic Inducible Fate Mapping (GIFM) to mark Gbx2-expressing precursors with fine temporal resolution and to subsequently track this lineage through embryogenesis. We then determined the terminal neuronal fate of the Gbx2 lineage in the adult Cb. Our analysis demonstrates that the Gbx2 lineage contributes to the Cb with marking over the course of five stages: Embryonic day 7.5 (E7.5) through E11.5. The Gbx2 lineage gives rise to Purkinje cells, granule neurons, and deep cerebellar neurons across these marking stages. Notably, the contribution of the Gbx2 lineage shifts as development proceeds with each marking stage producing a distinct profile of mature neurons in the adult Cb. These findings demonstrate the relationship between the temporal expression of Gbx2 and the terminal cell fate of neurons in the Cb. Based on these results, Gbx2 is critical to Cb development, not only for its well-defined role in positioning and maintaining the IsO, but also for guiding the development of Cb precursors and determining the identity of Cb neurons.
Collapse
Affiliation(s)
- Nellwyn Hagan
- Division of Biology and Medicine, Department of Neuroscience, Brown UniversityProvidence, RI, United States
| | - Juliana Guarente
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown UniversityProvidence, RI, United States
| | - Debra Ellisor
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown UniversityProvidence, RI, United States
| | - Mark Zervas
- Division of Biology and Medicine, Department of Neuroscience, Brown UniversityProvidence, RI, United States.,Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown UniversityProvidence, RI, United States.,Department of Neuroscience, AmgenCambridge, MA, United States
| |
Collapse
|
18
|
Hardy A, Benford D, Halldorsson T, Jeger MJ, Knutsen HK, More S, Naegeli H, Noteborn H, Ockleford C, Ricci A, Rychen G, Schlatter JR, Silano V, Solecki R, Turck D, Bresson JL, Dusemund B, Gundert-Remy U, Kersting M, Lambré C, Penninks A, Tritscher A, Waalkens-Berendsen I, Woutersen R, Arcella D, Court Marques D, Dorne JL, Kass GE, Mortensen A. Guidance on the risk assessment of substances present in food intended for infants below 16 weeks of age. EFSA J 2017; 15:e04849. [PMID: 32625502 PMCID: PMC7010120 DOI: 10.2903/j.efsa.2017.4849] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Following a request from the European Commission to EFSA, the EFSA Scientific Committee (SC) prepared a guidance for the risk assessment of substances present in food intended for infants below 16 weeks of age. In its approach to develop this guidance, the EFSA SC took into account, among others, (i) an exposure assessment based on infant formula as the only source of nutrition; (ii) knowledge of organ development in human infants, including the development of the gut, metabolic and excretory capacities, the brain and brain barriers, the immune system, the endocrine and reproductive systems; (iii) the overall toxicological profile of the substance identified through the standard toxicological tests, including critical effects; (iv) the relevance for the human infant of the neonatal experimental animal models used. The EFSA SC notes that during the period from birth up to 16 weeks, infants are expected to be exclusively fed on breast milk and/or infant formula. The EFSA SC views this period as the time where health-based guidance values for the general population do not apply without further considerations. High infant formula consumption per body weight is derived from 95th percentile consumption. The first weeks of life is the time of the highest relative consumption on a body weight basis. Therefore, when performing an exposure assessment, the EFSA SC proposes to use the high consumption value of 260 mL/kg bw per day. A decision tree approach is proposed that enables a risk assessment of substances present in food intended for infants below 16 weeks of age. The additional information needed when testing substances present in food for infants below 16 weeks of age and the approach to be taken for the risk assessment are on a case-by-case basis, depending on whether the substance is added intentionally to food and is systemically available.
Collapse
|
19
|
Hami J, Vafaei-Nezhad S, Ivar G, Sadeghi A, Ghaemi K, Mostafavizadeh M, Hosseini M. Altered expression and localization of synaptophysin in developing cerebellar cortex of neonatal rats due to maternal diabetes mellitus. Metab Brain Dis 2016; 31:1369-1380. [PMID: 27389246 DOI: 10.1007/s11011-016-9864-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 06/28/2016] [Indexed: 12/30/2022]
Abstract
There is sufficient evidence that diabetes during pregnancy is associated with a higher risk of neurodevelopmental anomalies including learning deficits, behavioral problems and motor dysfunctions in the offspring. Synaptophysin (SYP) is an integral membrane protein of synaptic vesicles and is considered as a marker for synaptogenesis and synaptic density. This study aimed to examine the effects of maternal diabetes in pregnancy on the expression and localization of SYP in the developing rat cerebellum. Wistar female rats were maintained diabetic from a week before pregnancy through parturition and male offspring was euthanized at postnatal day (P) 0, 7, and 14. The results revealed a significant down-regulation in the mRNA expression of SYP in the offspring born to diabetic animals at both P7 and P14 (P < 0.05 each). One week after birth, there was a significant reduction in the localization of SYP expression in the external granular (EGL) and in the molecular (ML) layers of neonates born to diabetic animals (P < 0.05 each). We also found a marked decrease in the expression of SYP in all of the cerebellar cortical layers of STZ-D group pups at P14 (P < 0.05 each). Moreover, our results revealed no significant changes in either expression or localization of SYP in insulin-treated group pups when compared with the controls (P ≥ 0.05 each). The present study demonstrated that maternal diabetes has adverse effects on the synaptogenesis in the offspring's cerebellum. Furthermore, the rigid maternal blood glucose control in the most cases normalized these negative impacts.
Collapse
Affiliation(s)
- Javad Hami
- Department of Anatomical Sciences, School of Medicine, Birjand University of Medical Sciences, Ghaffari St, Birjand, Iran
| | - Saeed Vafaei-Nezhad
- Department of Anatomical Sciences, School of Medicine, Birjand University of Medical Sciences, Ghaffari St, Birjand, Iran.
| | - Ghasem Ivar
- Department of Anatomical Sciences, School of Medicine, Birjand University of Medical Sciences, Ghaffari St, Birjand, Iran
| | - Akram Sadeghi
- Department of Anatomy and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Kazem Ghaemi
- Department of Neurosurgery, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Mehran Hosseini
- Department of Public Health, Deputy of Research and Technology, Research Centre of Experimental Medicine, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
20
|
Brossard-Racine M, Limperopoulos C. Normal Cerebellar Development by Qualitative and Quantitative MR Imaging. Neuroimaging Clin N Am 2016; 26:331-9. [DOI: 10.1016/j.nic.2016.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
21
|
Abstract
We aim to review the magnetic resonance imaging appearance of malformations of midbrain and hindbrain. These can be classified as predominantly cerebellar malformations, combined cerebellar and brain stem malformations, and predominantly brain stem malformations. The diagnostic criteria for the majority of these morphological malformations are based on neuroimaging findings. The predominantly cerebellar malformations include predominantly vermian hypoplasia seen in Dandy-Walker malformation and rhombencephalosynapsis, global cerebellar hypoplasia reported in lissencephaly and microlissencephaly, and unilateral cerebellar hypoplasia seen in PHACES, vanishing cerebellum, and cerebellar cleft. Cerebellar dysplasias are seen in Chudley-McCullough syndrome, associated with LAMA1 mutations and GPR56 mutations; Lhermitte-Duclos disease; and focal cerebellar dysplasias. Cerebellar hyperplasias are seen in megalencephaly-related syndromes and hemimegalencephaly with ipsilateral cerebellomegaly. Cerebellar and brain stem malformations include tubulinopathies, Joubert syndrome, cobblestone malformations, pontocerebellar hypoplasias, and congenital disorders of glycosylation type Ia. Predominantly brain stem malformations include congenital innervation dysgenesis syndrome, pontine tegmental cap dysplasia, diencephalic-mesencephalic junction dysplasia, disconnection syndrome, and pontine clefts.
Collapse
|
22
|
Wiethoff S, Arber C, Li A, Wray S, Houlden H, Patani R. Using human induced pluripotent stem cells to model cerebellar disease: hope and hype. J Neurogenet 2015; 29:95-102. [PMID: 25985846 PMCID: PMC4673530 DOI: 10.3109/01677063.2015.1053478] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/18/2015] [Indexed: 12/19/2022]
Abstract
The cerebellum forms a highly ordered and indispensible component of motor function within the adult neuraxis, consisting of several distinct cellular subtypes. Cerebellar disease, through a variety of genetic and acquired causes, results in the loss of function of defined subclasses of neurons, and remains a significant and untreatable health care burden. The scarcity of therapies in this arena can partially be explained by unresolved disease mechanisms due to inaccessibility of human cerebellar neurons in a relevant experimental context where initiating disease mechanisms could be functionally elucidated, or drug screens conducted. In this review we discuss the potential promise of human induced pluripotent stem cells (hiPSCs) for regenerative neurology, with a particular emphasis on in vitro modelling of cerebellar degeneration. We discuss progress made thus far using hiPSC-based models of neurodegeneration, noting the relatively slower pace of discovery made in modelling cerebellar dysfunction. We conclude by speculating how strategies attempting cerebellar differentiation from hiPSCs can be refined to allow the generation of accurate disease models. This in turn will permit a greater understanding of cerebellar pathophysiology to inform mechanistically rationalised therapies, which are desperately needed in this field.
Collapse
Affiliation(s)
- Sarah Wiethoff
- National Hospital for Neurology and Neurosurgery, UCL Institute of Neurology, London, UK
- Center for Neurology and Hertie Institute for Clinical Brain Research, Eberhard-Karls-University, Tübingen, Germany
| | - Charles Arber
- Department of Molecular Neuroscience and Queen Square Brain Bank, UCL Institute of Neurology, London, UK
| | - Abi Li
- Department of Molecular Neuroscience and Queen Square Brain Bank, UCL Institute of Neurology, London, UK
| | - Selina Wray
- Department of Molecular Neuroscience and Queen Square Brain Bank, UCL Institute of Neurology, London, UK
| | - Henry Houlden
- National Hospital for Neurology and Neurosurgery, UCL Institute of Neurology, London, UK
| | - Rickie Patani
- National Hospital for Neurology and Neurosurgery, UCL Institute of Neurology, London, UK
- Department of Molecular Neuroscience and Queen Square Brain Bank, UCL Institute of Neurology, London, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Euan MacDonald Centre for MND, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
23
|
Update on neuroimaging phenotypes of mid-hindbrain malformations. Neuroradiology 2014; 57:113-38. [DOI: 10.1007/s00234-014-1431-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 09/04/2014] [Indexed: 12/11/2022]
|
24
|
Multidimensional analysis of fetal posterior fossa in health and disease. THE CEREBELLUM 2014; 12:632-44. [PMID: 23553467 DOI: 10.1007/s12311-013-0470-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Fetal magnetic resonance imaging (MRI) is now routinely used to further investigate cerebellar malformations detected with ultrasound. However, the lack of 2D and 3D biometrics in the current literature hinders the detailed characterisation and classification of cerebellar anomalies. The main objectives of this fetal neuroimaging study were to provide normal posterior fossa growth trajectories during the second and third trimesters of pregnancy via semi-automatic segmentation of reconstructed fetal brain MR images and to assess common cerebellar malformations in comparison with the reference data. Using a 1.5-T MRI scanner, 143 MR images were obtained from 79 normal control and 53 fetuses with posterior fossa abnormalities that were grouped according to the severity of diagnosis on visual MRI inspections. All quantifications were performed on volumetric datasets, and supplemental outcome information was collected from the surviving infants. Normal growth trajectories of total brain, cerebellar, vermis, pons and fourth ventricle volumes showed significant correlations with 2D measurements and increased in second-order polynomial trends across gestation (Pearson r, p < 0.05). Comparison of normal controls to five abnormal cerebellum subgroups depicted significant alterations in volumes that could not be detected exclusively with 2D analysis (MANCOVA, p < 0.05). There were 15 terminations of pregnancy, 8 neonatal deaths, and a spectrum of genetic and neurodevelopmental outcomes in the assessed 24 children with cerebellar abnormalities. The given posterior fossa biometrics enhance the delineation of normal and abnormal cerebellar phenotypes on fetal MRI and confirm the advantages of utilizing advanced neuroimaging tools in clinical fetal research.
Collapse
|
25
|
The ciliary proteins Meckelin and Jouberin are required for retinoic acid-dependent neural differentiation of mouse embryonic stem cells. Differentiation 2014; 87:134-146. [PMID: 24613594 DOI: 10.1016/j.diff.2014.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 01/23/2014] [Accepted: 02/17/2014] [Indexed: 12/29/2022]
Abstract
The dysfunction of the primary cilium, a complex, evolutionarily conserved, organelle playing an important role in sensing and transducing cell signals, is the unifying pathogenetic mechanism of a growing number of diseases collectively termed "ciliopathies", typically characterized by multiorgan involvement. Developmental defects of the central nervous system (CNS) characterize a subset of ciliopathies showing clinical and genetic overlap, such as Joubert syndrome (JS) and Meckel syndrome (MS). Although several knock-out mice lacking a variety of ciliary proteins have shown the importance of primary cilia in the development of the brain and CNS-derived structures, developmental in vitro studies, extremely useful to unravel the role of primary cilia along the course of neural differentiation, are still missing. Mouse embryonic stem cells (mESCs) have been recently proven to mimic brain development, giving the unique opportunity to dissect the CNS differentiation process along its sequential steps. In the present study we show that mESCs express the ciliary proteins Meckelin and Jouberin in a developmentally-regulated manner, and that these proteins co-localize with acetylated tubulin labeled cilia located at the outer embryonic layer. Further, mESCs differentiating along the neuronal lineage activate the cilia-dependent sonic hedgehog signaling machinery, which is impaired in Meckelin knock-out cells but results unaffected in Jouberin-deficient mESCs. However, both lose the ability to acquire a neuronal phenotype. Altogether, these results demonstrate a pivotal role of Meckelin and Jouberin during embryonic neural specification and indicate mESCs as a suitable tool to investigate the developmental impact of ciliary proteins dysfunction.
Collapse
|
26
|
Gelpi E, Bombi JA, Martinez-Saez E, Caral L, Ribalta T. Prominent EMA 'dots' in tumour-induced Bergmann gliosis. Histopathology 2013; 64:445-52. [PMID: 24117995 DOI: 10.1111/his.12260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 08/15/2013] [Indexed: 11/29/2022]
Abstract
AIMS To describe an unusual pattern of epithelial membrane antigen (EMA) immunoreactivity in highly proliferative human Bergmann glia. METHODS AND RESULTS An immunohistochemical study was performed of postmortem cerebellar tissue from 18 adult patients with cerebellar damage of various aetiologies and 15 biopsies of diverse adult and paediatric cerebellar tumours. We observed marked proliferation of Bergmann glia with unusual prominent dot-like cytoplasmic EMA immunoreactivity in a case with extensive leptomeningeal sarcomatosis. Similar staining was not observed in association with other types of cerebellar pathology, except for other neoplastic conditions, such as leptomeningeal carcinomatosis, adult medulloblastoma, and pilocytic astrocytoma in children. At an ultrastructural level, the index case showed prominent endoplasmic reticulum with some intermediate filaments and lipofuscin granules, but no structures related to cilia or microvilli were observed. CONCLUSIONS We consider that prominent EMA dots in Bergmann glia might represent excessive activation induced by an overlying leptomeningeal tumour that stimulates the expression of early developmental antigens. This observation suggests modulation of the glial phenotype when exposed to a neoplastic microenvironment that, in turn, might influence the regenerative potential of Bergmann glia.
Collapse
Affiliation(s)
- Ellen Gelpi
- Neurological Tissue Bank of the Biobanc, Hospital Clinic, Insitut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | | | | | | |
Collapse
|
27
|
Oostland M, van Hooft J. The role of serotonin in cerebellar development. Neuroscience 2013; 248:201-12. [DOI: 10.1016/j.neuroscience.2013.05.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 05/16/2013] [Accepted: 05/17/2013] [Indexed: 01/09/2023]
|
28
|
Abstract
The cerebellum plays a role not only in motor control but also in motor learning and cognition. Joubert syndrome is a rare heterogeneous inherited genetic disorder characterized by ataxia, hypotonia, developmental delay, and at least one of the following features: neonatal respiratory disturbances or abnormal eye movement. The estimated frequency of Joubert syndrome in the United States is around 1 : 100 000. The term Joubert syndrome and related disorders (JSRD) has been recently coined to describe all disorders presenting with molar tooth sign on brain neuroimaging. Joubert syndrome is believed to be a representative of a new group of disorders named ciliopathies. The identification of seven causal genes (NPHP1, AHI1, CEP290, RPGRIP1L, TMEM67/MKS3, ARL13B, CC2D2A) has led to substantial progress in the understanding of the genetic basis of Joubert syndrome. The authors focus on clinical presentation of JSRD, differential diagnosis and molecular background.
Collapse
|
29
|
Joubert syndrome: brain and spinal cord malformations in genotyped cases and implications for neurodevelopmental functions of primary cilia. Acta Neuropathol 2012; 123:695-709. [PMID: 22331178 DOI: 10.1007/s00401-012-0951-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 01/25/2012] [Accepted: 01/27/2012] [Indexed: 12/12/2022]
Abstract
Joubert syndrome (JS) is an autosomal recessive ciliopathy characterized by hypotonia, ataxia, abnormal eye movements, and intellectual disability. The brain is malformed, with severe vermian hypoplasia, fourth ventriculomegaly, and "molar tooth" appearance of the cerebral and superior cerebellar peduncles visible as consistent features on neuroimaging. Neuropathological studies, though few, suggest that several other brain and spinal cord structures, such as the dorsal cervicomedullary junction, may also be affected in at least some patients. Genetically, JS is heterogeneous, with mutations in 13 genes accounting for approximately 50% of patients. Here, we compare neuropathologic findings in five subjects with JS, including four with defined mutations in OFD1 (2 siblings), RPGRIP1L, or TCTN2. Characteristic findings in all JS genotypes included vermian hypoplasia, fragmented dentate and spinal trigeminal nuclei, hypoplastic pontine and inferior olivary nuclei, and nondecussation of corticospinal tracts. Other common findings, seen in multiple genotypes but not all subjects, were dorsal cervicomedullary heterotopia, nondecussation of superior cerebellar peduncles, enlarged arcuate nuclei, hypoplastic reticular formation, hypoplastic medial lemnisci, and dorsal spinal cord disorganization. Thus, while JS exhibits significant neuropathologic as well as genetic heterogeneity, no genotype-phenotype correlations are apparent as yet. Our findings suggest that primary cilia are important for neural patterning, progenitor proliferation, cell migration, and axon guidance in the developing human brain and spinal cord.
Collapse
|
30
|
Armién AG, McRuer DL, Ruder MG, Wünschmann A. Purkinje Cell Heterotopy With Cerebellar Hypoplasia in Two Free-Living American Kestrels (Falco sparverius). Vet Pathol 2012; 50:182-7. [DOI: 10.1177/0300985812442690] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Two wild fledgling kestrels exhibited lack of motor coordination, postural reaction deficits, and abnormal propioception. At necropsy, the cerebellum and brainstem were markedly underdeveloped. Microscopically, there was Purkinje cells heterotopy, abnormal circuitry, and hypoplasia with defective foliation. Heterotopic neurons were identified as immature Purkinje cells by their size, location, immunoreactivity for calbindin D-28 K, and ultrastructural features. The authors suggest that this cerebellar abnormality was likely due to a disruption of molecular mechanisms that dictate Purkinje cell migration, placement, and maturation in early embryonic development. The etiology of this condition remains undetermined. Congenital central nervous system disorders have rarely been reported in birds.
Collapse
Affiliation(s)
- A. G. Armién
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - D. L. McRuer
- Wildlife Center of Virginia, Waynesboro, Virginia, USA
| | - M. G. Ruder
- Southeastern Cooperative Wildlife Disease Study, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - A. Wünschmann
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| |
Collapse
|
31
|
|
32
|
Key role of CXCL13/CXCR5 axis for cerebrospinal fluid B cell recruitment in pediatric OMS. J Neuroimmunol 2012; 243:81-8. [PMID: 22264765 DOI: 10.1016/j.jneuroim.2011.12.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 12/13/2011] [Accepted: 12/14/2011] [Indexed: 11/23/2022]
Abstract
To study aberrant B cell trafficking into the CSF in opsoclonus-myoclonus syndrome (OMS), chemoattractants CXCL13 and CXCL12, and B cell frequency and CXCR5 expression, were evaluated. CSF CXCL13 concentration and the CSF/serum ratio were higher in untreated OMS than controls, related directly to OMS severity and inversely to OMS duration, and correlated with CSF B cell frequency and oligoclonal bands. CXCL12 showed the opposite pattern. Selective accumulation of CXCR5+ memory B cells in CSF was found. In ACTH-treated OMS, CXCL13, but not CXCL12, was lower. These data implicate the chemokine/chemoreceptor pair CXCL13/CXR5 in B cell recruitment to the CNS in OMS. CXCL13 and CXCL12 may serve as reciprocal biomarkers of disease activity, but CXCL13 also had utility as a treatment biomarker.
Collapse
|
33
|
Abstract
The cerebellum undergoes a protracted development, making it particularly vulnerable to a broad spectrum of developmental events. Acquired destructive and hemorrhagic insults may also occur. The main steps of cerebellar development are reviewed. The normal imaging patterns of the cerebellum in prenatal ultrasound and magnetic resonance imaging (MRI) are described with emphasis on the limitations of these modalities. Because of confusion in the literature regarding the terminology used for cerebellar malformations, some terms (agenesis, hypoplasia, dysplasia, and atrophy) are clarified. Three main pathologic settings are considered and the main diagnoses that can be suggested are described: retrocerebellar fluid enlargement with normal or abnormal biometry (Dandy-Walker malformation, Blake pouch cyst, vermian agenesis), partially or globally decreased cerebellar biometry (cerebellar hypoplasia, agenesis, rhombencephalosynapsis, ischemic and/or hemorrhagic damage), partially or globally abnormal cerebellar echogenicity (ischemic and/or hemorrhagic damage, cerebellar dysplasia, capillary telangiectasia). The appropriate timing for performing MRI is also discussed.
Collapse
Affiliation(s)
- Catherine Garel
- Service de Radiologie, Hôpital d'Enfants Armand-Trousseau, Paris, France.
| | | | | |
Collapse
|
34
|
Urkasemsin G, Linder KE, Bell JS, de Lahunta A, Olby NJ. Mapping of Purkinje Neuron Loss and Polyglucosan Body Accumulation in Hereditary Cerebellar Degeneration in Scottish Terriers. Vet Pathol 2011; 49:852-9. [DOI: 10.1177/0300985811412622] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
A hereditary cerebellar degenerative disorder has emerged in Scottish Terriers. The aims of this study were to describe and quantify polyglucosan body accumulation and quantify Purkinje neurons in the cerebellum of affected and control dogs. The brains of 6 affected Scottish Terriers ranging in age from 8 to 15 years and 8 age-matched control dogs were examined histopathologically. Counts of Purkinje neurons and polyglucosan bodies were performed in control and affected dogs on cerebellar sections stained with periodic acid–Schiff. Affected dogs showed a significant loss of Purkinje neurons compared with control dogs (vermis: P < .0001; hemisphere: P = .0104). The degeneration was significantly more pronounced dorsally than ventrally ( P < .0001). There were significantly more polyglucosan bodies in the ventral half of the vermis when compared with the dorsal half ( P < .0001) in affected dogs. In addition, there were more polyglucosan bodies in the ventral half of the vermis in affected dogs than in control dogs ( P = .0005). Polyglucosan bodies in all affected dogs stained positively with toluidine blue and alcian blue. Immunohistochemically, polyglucosan bodies in affected dogs were positive for neurofilament 200 kD and ubiquitin and negative for glial fibrillary acidic protein, synaptophysin, neurospecific enolase, vimentin, and S100; the bodies were negative for all antigens in control dogs. Ultrastructurally, polyglucosan bodies in 1 affected dog were non–membrane-bound, amorphous structures with a dense core. This study demonstrates significant Purkinje cell loss and increased polyglucosan bodies in the cerebellum of affected Scottish Terriers.
Collapse
Affiliation(s)
- G. Urkasemsin
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
| | - K. E. Linder
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
| | - J. S. Bell
- Department of Clinical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA
| | - A. de Lahunta
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY
| | - N. J. Olby
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
- Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
| |
Collapse
|
35
|
Alcaraz WA, Chen E, Valdes P, Kim E, Lo YH, Vo J, Hamilton BA. Modifier genes and non-genetic factors reshape anatomical deficits in Zfp423-deficient mice. Hum Mol Genet 2011; 20:3822-30. [PMID: 21729880 DOI: 10.1093/hmg/ddr300] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Development of neural circuitry depends on the integration of signaling pathways to coordinate specification, proliferation and differentiation of cell types in the right number, in the right place, at the right time. Zinc finger protein 423 (Zfp423), a 30-zinc finger transcription factor, forms alternate complexes with components of several developmental signaling pathways, suggesting it as a point of signal integration during brain development. We previously showed that mice lacking Zfp423 have reduced proliferation of cerebellar precursor cells, resulting in complete loss of vermis and variable hypoplasia of cerebellar hemispheres. Here, we show that Zfp423(-/-) hemisphere malformations are shaped by both genetic and non-genetic factors, producing distinct phenotype distributions in different inbred genetic backgrounds. In genetic mapping studies, we identify four additive modifier loci (Amzn1-4) and seven synthetically interacting loci (Smzn1.1-3.1) that together explain approximately one-third of the phenotypic variance. Strain-specific sequence polymorphism and expression data provide a reduced list of functional variant candidate genes at each modifier locus. Environmental covariates add only modest explanatory power, suggesting an additional stochastic component. These results provide a comprehensive analysis of sources of phenotype variation in a model of hindbrain malformation.
Collapse
Affiliation(s)
- Wendy A Alcaraz
- Biomedical Sciences Graduate Program,, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0644, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Pal S, Gupta R, Kim H, Wickramasinghe P, Baubet V, Showe LC, Dahmane N, Davuluri RV. Alternative transcription exceeds alternative splicing in generating the transcriptome diversity of cerebellar development. Genome Res 2011; 21:1260-72. [PMID: 21712398 DOI: 10.1101/gr.120535.111] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Despite our growing knowledge that many mammalian genes generate multiple transcript variants that may encode functionally distinct protein isoforms, the transcriptomes of various tissues and their developmental stages are poorly defined. Identifying the transcriptome and its regulation in a cell/tissue is the key to deciphering the cell/tissue-specific functions of a gene. We built a genome-wide inventory of noncoding and protein-coding transcripts (transcriptomes), their promoters (promoteromes) and histone modification states (epigenomes) for developing, and adult cerebella using integrative massive-parallel sequencing and bioinformatics approach. The data consists of 61,525 (12,796 novel) distinct mRNAs transcribed by 29,589 (4792 novel) promoters corresponding to 15,669 protein-coding and 7624 noncoding genes. Importantly, our results show that the transcript variants from a gene are predominantly generated using alternative transcriptional rather than splicing mechanisms, highlighting alternative promoters and transcriptional terminations as major sources of transcriptome diversity. Moreover, H3K4me3, and not H3K27me3, defined the use of alternative promoters, and we identified a combinatorial role of H3K4me3 and H3K27me3 in regulating the expression of transcripts, including transcript variants of a gene during development. We observed a strong bias of both H3K4me3 and H3K27me3 for CpG-rich promoters and an exponential relationship between their enrichment and corresponding transcript expression. Furthermore, the majority of genes associated with neurological diseases expressed multiple transcripts through alternative promoters, and we demonstrated aberrant use of alternative promoters in medulloblastoma, cancer arising in the cerebellum. The transcriptomes of developing and adult cerebella presented in this study emphasize the importance of analyzing gene regulation and function at the isoform level.
Collapse
Affiliation(s)
- Sharmistha Pal
- Center for Systems and Computational Biology, The Wistar Institute, Philadelphia, Pennsylvania 19019, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Meyer U, Feldon J, Dammann O. Schizophrenia and autism: both shared and disorder-specific pathogenesis via perinatal inflammation? Pediatr Res 2011; 69:26R-33R. [PMID: 21289540 PMCID: PMC3086802 DOI: 10.1203/pdr.0b013e318212c196] [Citation(s) in RCA: 254] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Prenatal exposure to infection and subsequent inflammatory responses have been implicated in the etiology of schizophrenia and autism. In this review, we summarize current evidence from human and animal studies supporting the hypothesis that the pathogenesis of these two disorders is linked via exposure to inflammation at early stages of development. Moreover, we propose a hypothetical model in which inflammatory mechanisms may account for multiple shared and disorder-specific pathological characteristics of both entities. In essence, our model suggests that acute neuroinflammation during early fetal development may be relevant for the induction of psychopathological and neuropathological features shared by schizophrenia and autism, whereas postacute latent and persistent inflammation may contribute to schizophrenia- and autism-specific phenotypes, respectively.
Collapse
Affiliation(s)
- Urs Meyer
- Laboratory of Behavioural Neurobiology, Swiss Federal Institute of Technology (ETH) Zurich, 8603 Schwerzenbach, Switzerland.
| | | | | |
Collapse
|
38
|
Davare MA, Saneyoshi T, Soderling TR. Calmodulin-kinases regulate basal and estrogen stimulated medulloblastoma migration via Rac1. J Neurooncol 2010; 104:65-82. [PMID: 21107644 DOI: 10.1007/s11060-010-0472-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Accepted: 11/12/2010] [Indexed: 01/05/2023]
Abstract
Medulloblastoma is a highly prevalent pediatric central nervous system malignancy originating in the cerebellum, with a strong propensity for metastatic migration to the leptomeninges, which greatly increases mortality. While numerous investigations are focused on the molecular mechanisms of medulloblastoma histogenesis, the signaling pathways regulating migration are still poorly understood. Medulloblastoma likely arises from aberrant proliferative signaling in cerebellar granule precursor cells during development, and estrogen is a morphogen that promotes medulloblastoma cell migration. It has been previously shown that the calcium/calmodulin activated kinase kinase (CaMKK) pathway promotes cerebellar granule precursor migration and differentiation during normal cerebellar development via CaMKIV. Here we investigate the regulatory role of the CaMKK pathway in migration of the human medulloblastoma DAOY and cerebellar granule cells. Using pharmacological inhibitors and dominant negative approaches, we demonstrate that the CaMKK/CaMKI cascade regulates basal medulloblastoma cell migration via Rac1, in part by activation of the RacGEF, βPIX. Additionally, pharmacological inhibition of CaMKK blocks both the estrogen induced Rac1 activation and medulloblastoma migration. The CaMKK signaling module described here is one of the first reported calcium regulated pathways that modulates medulloblastoma migration. Since tumor dissemination requires cell migration to ectopic sites, this CaMKK pathway may be a putative therapeutic target to limit medulloblastoma metastasis.
Collapse
Affiliation(s)
- Monika A Davare
- Vollum Institute and Knight Cancer Institute, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| | | | | |
Collapse
|
39
|
Posterior fossa malformations: main features and limits in prenatal diagnosis. Pediatr Radiol 2010; 40:1038-45. [PMID: 20432023 DOI: 10.1007/s00247-010-1617-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 01/06/2010] [Indexed: 12/11/2022]
Abstract
Posterior fossa (PF) malformations are commonly observed during prenatal screening. Their understanding requires knowledge of the main steps of PF development and knowledge of normal patterns in US and MR imaging. The vast majority of PF malformations can be strongly suspected by acquiring a midline sagittal slice and a transverse slice and by systematically scrutinizing the elements of the PF: cerebellar vermis, hemispheres, brainstem, fourth ventricle, PF fluid spaces and tentorium. Analysis of cerebellar echogenicity and biometry is also useful. This review explains how to approach the diagnosis of the main PF malformations by performing these two slices and answering six key questions about the elements of the PF. The main imaging characteristics of PF malformations are also reviewed.
Collapse
|
40
|
Juranek J, Dennis M, Cirino PT, El-Messidi L, Fletcher JM. The cerebellum in children with spina bifida and Chiari II malformation: Quantitative volumetrics by region. CEREBELLUM (LONDON, ENGLAND) 2010; 9:240-8. [PMID: 20143197 PMCID: PMC3046026 DOI: 10.1007/s12311-010-0157-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Few volumetric MRI studies of the entire cerebellum have been published; even less quantitative information is available in patients with hindbrain malformations, including the Chiari II malformation which is ubiquitous in patients with spina bifida meningomyelocele (SBM). In the present study, regional volumetric analyses of the cerebellum were conducted in children with SBM/Chiari II and typically developing (TD) children. Total cerebellar volume was significantly reduced in the SBM group relative to the TD group. After correcting for total cerebellum volume, and relative to the TD group, the posterior lobe was significantly reduced in SBM, the corpus medullare was not different, and the anterior lobe was significantly enlarged. Children with thoracic level lesions had smaller cerebellar volumes relative to those with lumbar/sacral lesions, who had smaller volumes compared to TD children. The reduction in cerebellar volume in the group with SBM represents not a change in linear scaling but rather a reconfiguration involving anterior lobe enlargement and posterior lobe reduction.
Collapse
Affiliation(s)
- Jenifer Juranek
- Department of Pediatrics, Children's Learning Institute, University of Texas Health Science Center at Houston, Houston, 77030, USA.
| | | | | | | | | |
Collapse
|