1
|
Imamura M, Yoshino M, Kawasaki H. Investigation of the development and evolution of the mammalian cerebrum using gyrencephalic ferrets. Eur J Cell Biol 2024; 103:151466. [PMID: 39546916 DOI: 10.1016/j.ejcb.2024.151466] [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: 08/31/2024] [Revised: 11/02/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
Mammalian brains have evolved a neocortex, which has diverged in size and morphology in different species over the course of evolution. In some mammals, a substantial increase in the number of neurons and glial cells resulted in the expansion and folding of the cerebrum, and it is believed that these evolutionary changes contributed to the acquisition of higher cognitive abilities in mammals. However, their underlying molecular and cellular mechanisms remain insufficiently elucidated. A major difficulty in addressing these mechanisms stemmed from the lack of appropriate animal models, as conventional experimental animals such as mice and rats have small brains without structurally obvious folds. Therefore, researchers including us have focused on using ferrets instead of mice and rats. Ferrets are domesticated carnivorous mammals with a gyrencephalic cerebrum, and, notably, they are amenable to genetic manipulations including in utero electroporation to knock out genes in the cerebrum. In this review, we highlight recent research into the mechanisms underlying the development and evolution of cortical folds using ferrets.
Collapse
Affiliation(s)
- Masanori Imamura
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan; Sapiens Life Sciences, Evolution and Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Mayuko Yoshino
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan; Sapiens Life Sciences, Evolution and Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan; Sapiens Life Sciences, Evolution and Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan.
| |
Collapse
|
2
|
Manning LK, Winkenwerder E, Baskind L, Eager KLM, Willet CE, Porebski B, O'Rourke BA, Tammen I, Gimeno M, Pinczowski P. A novel missense variant in the RELN gene in sheep with lissencephaly and cerebellar hypoplasia. Vet Pathol 2024:3009858241283501. [PMID: 39394905 DOI: 10.1177/03009858241283501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2024]
Abstract
Lissencephaly and cerebellar hypoplasia (LCH) represents a spectrum of congenital developmental malformations of the cerebral cortex and cerebellum, mostly occurring as inherited conditions caused by variants in an increasingly recognized number of genes. LCH has been identified in three Dorset-cross lambs with congenital neurological signs in Australia. Lambs were unable to walk and had reduced vision, and one lamb developed a hypermetric gait and intention tremors. Grossly, the lambs had diffuse pachygyria with reduction in white matter, mild bilateral ventriculomegaly of the lateral ventricles, and a markedly hypoplastic cerebellum. Histologically, there was disorganization of neurons within the cerebral cortex and hippocampus. The cerebellar vermis had disorganized, thin, and hypocellular gray matter with frequent ectopic Purkinje cells, while identifiable folia were largely absent within the hemispheres. Luxol fast blue stain and glial fibrillary acidic protein, neuronal nuclear protein, synaptophysin, and neuron-specific enolase immunohistochemistry confirmed the thickened, disorganized cerebral cortical gray matter and reduced white matter. Within the cerebellum, immunohistochemistry demonstrated marked dysplasia. Whole-genome sequencing analysis and prediction of variant effects identified a missense variant of interest in the candidate gene reelin (RELN; NC_040255.1:g.50288685C>T; NM_001306121.1:c.7088G>A; NP_001293050.1:p.(R2363H)) with a deleterious Sorting Intolerant from Tolerant (SIFT) score. Sanger sequencing identified that the variant segregated with LCH disease in the 3 affected individuals, their sire, and 6 unaffected flock members. The NP_001293050.1: p.(R2363H) substitution is predicted to decrease the stability of the protein (ΔΔG = -1.55 kcal/mol). Pathological and genetic findings are consistent with previously described phenotypes of RELN variants in Churra sheep, dogs, and humans.
Collapse
Affiliation(s)
- Leah K Manning
- Elizabeth Macarthur Agricultural Institute, NSW Department of Primary Industries and Regional Development, Menangle, NSW, Australia
- The University of Sydney, Camden, NSW, Australia
| | - Emily Winkenwerder
- Elizabeth Macarthur Agricultural Institute, NSW Department of Primary Industries and Regional Development, Menangle, NSW, Australia
- The University of Sydney, Camden, NSW, Australia
| | - Louise Baskind
- South East Local Land Services, Braidwood, NSW, Australia
| | - Katie L M Eager
- Elizabeth Macarthur Agricultural Institute, NSW Department of Primary Industries and Regional Development, Menangle, NSW, Australia
- The University of Sydney, Camden, NSW, Australia
| | - Cali E Willet
- The University of Sydney, Camperdown, NSW, Australia
| | - Ben Porebski
- Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Brendon A O'Rourke
- Elizabeth Macarthur Agricultural Institute, NSW Department of Primary Industries and Regional Development, Menangle, NSW, Australia
| | - Imke Tammen
- The University of Sydney, Camden, NSW, Australia
| | - Marina Gimeno
- Elizabeth Macarthur Agricultural Institute, NSW Department of Primary Industries and Regional Development, Menangle, NSW, Australia
- The University of Sydney, Camden, NSW, Australia
| | - Pedro Pinczowski
- Elizabeth Macarthur Agricultural Institute, NSW Department of Primary Industries and Regional Development, Menangle, NSW, Australia
| |
Collapse
|
3
|
Zhu X, Ma S, Wong WH. Genetic effects of sequence-conserved enhancer-like elements on human complex traits. Genome Biol 2024; 25:1. [PMID: 38167462 PMCID: PMC10759394 DOI: 10.1186/s13059-023-03142-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND The vast majority of findings from human genome-wide association studies (GWAS) map to non-coding sequences, complicating their mechanistic interpretations and clinical translations. Non-coding sequences that are evolutionarily conserved and biochemically active could offer clues to the mechanisms underpinning GWAS discoveries. However, genetic effects of such sequences have not been systematically examined across a wide range of human tissues and traits, hampering progress to fully understand regulatory causes of human complex traits. RESULTS Here we develop a simple yet effective strategy to identify functional elements exhibiting high levels of human-mouse sequence conservation and enhancer-like biochemical activity, which scales well to 313 epigenomic datasets across 106 human tissues and cell types. Combined with 468 GWAS of European (EUR) and East Asian (EAS) ancestries, these elements show tissue-specific enrichments of heritability and causal variants for many traits, which are significantly stronger than enrichments based on enhancers without sequence conservation. These elements also help prioritize candidate genes that are functionally relevant to body mass index (BMI) and schizophrenia but were not reported in previous GWAS with large sample sizes. CONCLUSIONS Our findings provide a comprehensive assessment of how sequence-conserved enhancer-like elements affect complex traits in diverse tissues and demonstrate a generalizable strategy of integrating evolutionary and biochemical data to elucidate human disease genetics.
Collapse
Affiliation(s)
- Xiang Zhu
- Department of Statistics, The Pennsylvania State University, 326 Thomas Building, University Park, 16802, PA, USA.
- Huck Institutes of the Life Sciences, The Pennsylvania State University, 201 Huck Life Sciences Building, University Park, 16802, PA, USA.
- Department of Statistics, Stanford University, 390 Jane Stanford Way, Stanford, 94305, CA, USA.
| | - Shining Ma
- Department of Statistics, Stanford University, 390 Jane Stanford Way, Stanford, 94305, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, 1265 Welch Road MC5464, Stanford, 94305, CA, USA
| | - Wing Hung Wong
- Department of Statistics, Stanford University, 390 Jane Stanford Way, Stanford, 94305, CA, USA.
- Department of Biomedical Data Science, Stanford University School of Medicine, 1265 Welch Road MC5464, Stanford, 94305, CA, USA.
| |
Collapse
|
4
|
Simoes C, Graña M, Rodriguez S, Baltar Yanes F, Tapié A, Dell’Oca N, Naya H, Raggio V, Spangenberg L. Novel frameshift mutation in LIS1 gene is a probable cause of lissencephaly: a case report. BMC Pediatr 2022; 22:545. [PMID: 36100855 PMCID: PMC9472359 DOI: 10.1186/s12887-022-03595-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 09/02/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Lissencephaly (LIS) is a cortical malformation, characterized by smooth or nearly smooth cerebral surface and a shortage of gyral and sulcal development, which is caused by deficient neuronal migration during embryogenesis. Neuronal migration involves many gene products, among which is the product of the PAFAH1B1 gene, associated with this disease. LIS is a rare disease, characterized by low population frequency, and with non-specific clinical symptoms such as early epilepsy, developmental delay or cerebral palsy-like motor problems. Given that high-throughput sequencing techniques have been improving diagnosis, we have chosen this technique for addressing this patient.
Case presentation
We present the case of a seven years old male patient with an undiagnosed rare disease, with non-specific clinical symptoms possibly compatible with lissencephaly.
The patient was enrolled in a study that included the sequencing of his whole genome. Sequence data was analyzed following a bioinformatic pipeline. The variants obtained were annotated and then subjected to different filters for prioritization. Also mitochondrial genome was analyzed. A novel candidate frameshift insertion in known PAFAH1B1 gene was found, explaining the index case phenotype. The assessment through in silico tools reported that it causes nonsense mediated mechanisms and that it is damaging with high confidence scores. The insertion causes a change in the reading frame, and produces a premature stop codon, severely affecting the protein function and probably the silencing of one allele. The healthy mother did not carry the mutation, and the unaffected father was not available for analysis.
Conclusions
Through this work we found a novel de novo mutation in LIS1/PAFAH1B1 gene, as a likely cause of a rare disease in a young boy with non-specific clinical symptoms. The mutation found correlates with the phenotype studied since the loss of function in the gene product has already been described in this condition. Since there are no other variants in the PAFAH1B1 gene with low population frequency and due to family history, a de novo disease mechanism is proposed.
Collapse
|
5
|
Maillard C, Roux CJ, Charbit-Henrion F, Steffann J, Laquerriere A, Quazza F, Buisson NB. Tubulin mutations in human neurodevelopmental disorders. Semin Cell Dev Biol 2022; 137:87-95. [PMID: 35915025 DOI: 10.1016/j.semcdb.2022.07.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/22/2022] [Accepted: 07/22/2022] [Indexed: 10/16/2022]
Abstract
Mutations causing dysfunction of tubulins and microtubule-associated proteins, also known as tubulinopathies, are a group of recently described entities that lead to complex brain malformations. Anatomical and functional consequences of the disruption of tubulins include microcephaly, combined with abnormal corticogenesis due to impaired migration or lamination and abnormal growth cone dynamics of projecting and callosal axons. Key imaging features of tubulinopathies are characterized by three major patterns of malformations of cortical development (MCD): lissencephaly, microlissencephaly, and dysgyria. Additional distinctive MRI features include dysmorphism of the basal ganglia, midline commissural structure hypoplasia or agenesis, and cerebellar and brainstem hypoplasia. Tubulinopathies can be diagnosed as early as 21-24 gestational weeks using imaging and neuropathology, with possible extreme microlissencephaly with an extremely thin cortex, lissencephaly with either thick or thin/intermediate cortex, and dysgyria combined with cerebellar hypoplasia, pons hypoplasia and corpus callosum dysgenesis. More than 100 MCD-associated mutations have been reported in TUBA1A, TUBB2B, or TUBB3 genes, whereas fewer than ten are known in other genes such TUBB2A, TUBB or TUBG1. Although these mutations are scattered along the α- and β-tubulin sequences, recurrent mutations are consistently associated with almost identical cortical dysgenesis. Much of the evidence supports that these mutations alter the dynamic properties and functions of microtubules in several fashions. These include diminishing the abundance of functional tubulin heterodimers, altering GTP binding, altering longitudinal and lateral protofilament interactions, and impairing microtubule interactions with kinesin and/or dynein motors or with MAPs. In this review we discuss the recent advances in our understanding of the effects of mutations of tubulins and microtubule-associated proteins on human brain development and the pathogenesis of malformations of cortical development.
Collapse
Affiliation(s)
- Camille Maillard
- Université de Paris, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, F-75015 Paris, France; Université de Paris, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, F-75014 Paris, France
| | - Charles Joris Roux
- Pediatric Radiology, Necker Enfants Malades University Hospital, Université de Paris, Paris, France
| | - Fabienne Charbit-Henrion
- Université de Paris, Sorbonne Paris Cité, Imagine INSERM UMR1163, Service de Génétique Moléculaire, Groupe hospitalier Necker-Enfants Malades, AP-HP, France
| | - Julie Steffann
- Université de Paris, Sorbonne Paris Cité, Imagine INSERM UMR1163, Service de Génétique Moléculaire, Groupe hospitalier Necker-Enfants Malades, AP-HP, France
| | - Annie Laquerriere
- Pathology Laboratory, Rouen University Hospital, Rouen, France; NeoVasc Region-Inserm Team ERI28, Laboratory of Microvascular Endothelium and Neonate Brain Lesions, Institute of Research for Innovation in Biomedicine, University of Rouen, Rouen, France
| | - Floriane Quazza
- Pediatric Neurology, Necker Enfants Malades University Hospital, Université de Paris, Paris, France
| | - Nadia Bahi Buisson
- Université de Paris, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, F-75015 Paris, France; Université de Paris, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, F-75014 Paris, France; Pediatric Neurology, Necker Enfants Malades University Hospital, Université de Paris, Paris, France.
| |
Collapse
|
6
|
Hansen AH, Pauler FM, Riedl M, Streicher C, Heger A, Laukoter S, Sommer C, Nicolas A, Hof B, Tsai LH, Rülicke T, Hippenmeyer S. Tissue-Wide Effects Override Cell-Intrinsic Gene Function in Radial Neuron Migration. OXFORD OPEN NEUROSCIENCE 2022; 1:kvac009. [PMID: 38596707 PMCID: PMC10939316 DOI: 10.1093/oons/kvac009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/26/2022] [Accepted: 05/15/2022] [Indexed: 04/11/2024]
Abstract
The mammalian neocortex is composed of diverse neuronal and glial cell classes that broadly arrange in six distinct laminae. Cortical layers emerge during development and defects in the developmental programs that orchestrate cortical lamination are associated with neurodevelopmental diseases. The developmental principle of cortical layer formation depends on concerted radial projection neuron migration, from their birthplace to their final target position. Radial migration occurs in defined sequential steps, regulated by a large array of signaling pathways. However, based on genetic loss-of-function experiments, most studies have thus far focused on the role of cell-autonomous gene function. Yet, cortical neuron migration in situ is a complex process and migrating neurons traverse along diverse cellular compartments and environments. The role of tissue-wide properties and genetic state in radial neuron migration is however not clear. Here we utilized mosaic analysis with double markers (MADM) technology to either sparsely or globally delete gene function, followed by quantitative single-cell phenotyping. The MADM-based gene ablation paradigms in combination with computational modeling demonstrated that global tissue-wide effects predominate cell-autonomous gene function albeit in a gene-specific manner. Our results thus suggest that the genetic landscape in a tissue critically affects the overall migration phenotype of individual cortical projection neurons. In a broader context, our findings imply that global tissue-wide effects represent an essential component of the underlying etiology associated with focal malformations of cortical development in particular, and neurological diseases in general.
Collapse
Affiliation(s)
- Andi H Hansen
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Florian M Pauler
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Michael Riedl
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Carmen Streicher
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Anna Heger
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Susanne Laukoter
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Christoph Sommer
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Armel Nicolas
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Björn Hof
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Li Huei Tsai
- Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
| | - Thomas Rülicke
- Department of Biomedical Sciences, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Simon Hippenmeyer
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| |
Collapse
|
7
|
Schaaf ZA, Tat L, Cannizzaro N, Panoutsopoulos AA, Green R, Rülicke T, Hippenmeyer S, Zarbalis KS. WDFY3 mutation alters laminar position and morphology of cortical neurons. Mol Autism 2022; 13:27. [PMID: 35733184 PMCID: PMC9219247 DOI: 10.1186/s13229-022-00508-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 06/09/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Proper cerebral cortical development depends on the tightly orchestrated migration of newly born neurons from the inner ventricular and subventricular zones to the outer cortical plate. Any disturbance in this process during prenatal stages may lead to neuronal migration disorders (NMDs), which can vary in extent from focal to global. Furthermore, NMDs show a substantial comorbidity with other neurodevelopmental disorders, notably autism spectrum disorders (ASDs). Our previous work demonstrated focal neuronal migration defects in mice carrying loss-of-function alleles of the recognized autism risk gene WDFY3. However, the cellular origins of these defects in Wdfy3 mutant mice remain elusive and uncovering it will provide critical insight into WDFY3-dependent disease pathology. METHODS Here, in an effort to untangle the origins of NMDs in Wdfy3lacZ mice, we employed mosaic analysis with double markers (MADM). MADM technology enabled us to genetically distinctly track and phenotypically analyze mutant and wild-type cells concomitantly in vivo using immunofluorescent techniques. RESULTS We revealed a cell autonomous requirement of WDFY3 for accurate laminar positioning of cortical projection neurons and elimination of mispositioned cells during early postnatal life. In addition, we identified significant deviations in dendritic arborization, as well as synaptic density and morphology between wild type, heterozygous, and homozygous Wdfy3 mutant neurons in Wdfy3-MADM reporter mice at postnatal stages. LIMITATIONS While Wdfy3 mutant mice have provided valuable insight into prenatal aspects of ASD pathology that remain inaccessible to investigation in humans, like most animal models, they do not a perfectly replicate all aspects of human ASD biology. The lack of human data makes it indeterminate whether morphological deviations described here apply to ASD patients or some of the other neurodevelopmental conditions associated with WDFY3 mutation. CONCLUSIONS Our genetic approach revealed several cell autonomous requirements of WDFY3 in neuronal development that could underlie the pathogenic mechanisms of WDFY3-related neurodevelopmental conditions. The results are also consistent with findings in other ASD animal models and patients and suggest an important role for WDFY3 in regulating neuronal function and interconnectivity in postnatal life.
Collapse
Affiliation(s)
- Zachary A Schaaf
- University of California at Davis, Department of Pathology and Laboratory Medicine, Sacramento, CA, 95817, USA
- Shriners Hospitals for Children Northern California, Sacramento, CA, 95817, USA
| | - Lyvin Tat
- University of California at Davis, Department of Pathology and Laboratory Medicine, Sacramento, CA, 95817, USA
| | - Noemi Cannizzaro
- University of California at Davis, Department of Pathology and Laboratory Medicine, Sacramento, CA, 95817, USA
| | - Alexios A Panoutsopoulos
- Shriners Hospitals for Children Northern California, Sacramento, CA, 95817, USA
- University of California at Davis, Department of Physiology and Membrane Biology, Sacramento, CA, 95817, USA
| | - Ralph Green
- University of California at Davis, Department of Pathology and Laboratory Medicine, Sacramento, CA, 95817, USA
| | - Thomas Rülicke
- Department of Biomedical Sciences, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Simon Hippenmeyer
- Institute of Science and Technology Austria, Am Campus 1, 3400, Klosterneuburg, Austria
| | - Konstantinos S Zarbalis
- University of California at Davis, Department of Pathology and Laboratory Medicine, Sacramento, CA, 95817, USA.
- Shriners Hospitals for Children Northern California, Sacramento, CA, 95817, USA.
- UC Davis MIND Institute, Sacramento, CA, 95817, USA.
| |
Collapse
|
8
|
Shinmyo Y, Hamabe-Horiike T, Saito K, Kawasaki H. Investigation of the Mechanisms Underlying the Development and Evolution of the Cerebral Cortex Using Gyrencephalic Ferrets. Front Cell Dev Biol 2022; 10:847159. [PMID: 35386196 PMCID: PMC8977464 DOI: 10.3389/fcell.2022.847159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
The mammalian cerebral cortex has changed significantly during evolution. As a result of the increase in the number of neurons and glial cells in the cerebral cortex, its size has markedly expanded. Moreover, folds, called gyri and sulci, appeared on its surface, and its neuronal circuits have become much more complicated. Although these changes during evolution are considered to have been crucial for the acquisition of higher brain functions, the mechanisms underlying the development and evolution of the cerebral cortex of mammals are still unclear. This is, at least partially, because it is difficult to investigate these mechanisms using mice only. Therefore, genetic manipulation techniques for the cerebral cortex of gyrencephalic carnivore ferrets were developed recently. Furthermore, gene knockout was achieved in the ferret cerebral cortex using the CRISPR/Cas9 system. These techniques enabled molecular investigations using the ferret cerebral cortex. In this review, we will summarize recent findings regarding the mechanisms underlying the development and evolution of the mammalian cerebral cortex, mainly focusing on research using ferrets.
Collapse
Affiliation(s)
- Yohei Shinmyo
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Toshihide Hamabe-Horiike
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kengo Saito
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
9
|
Freeman-Cook K, Hoffman RL, Miller N, Almaden J, Chionis J, Zhang Q, Eisele K, Liu C, Zhang C, Huser N, Nguyen L, Costa-Jones C, Niessen S, Carelli J, Lapek J, Weinrich SL, Wei P, McMillan E, Wilson E, Wang TS, McTigue M, Ferre RA, He YA, Ninkovic S, Behenna D, Tran KT, Sutton S, Nagata A, Ornelas MA, Kephart SE, Zehnder LR, Murray B, Xu M, Solowiej JE, Visswanathan R, Boras B, Looper D, Lee N, Bienkowska JR, Zhu Z, Kan Z, Ding Y, Mu XJ, Oderup C, Salek-Ardakani S, White MA, VanArsdale T, Dann SG. Expanding control of the tumor cell cycle with a CDK2/4/6 inhibitor. Cancer Cell 2021; 39:1404-1421.e11. [PMID: 34520734 DOI: 10.1016/j.ccell.2021.08.009] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 06/03/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022]
Abstract
The CDK4/6 inhibitor, palbociclib (PAL), significantly improves progression-free survival in HR+/HER2- breast cancer when combined with anti-hormonals. We sought to discover PAL resistance mechanisms in preclinical models and through analysis of clinical transcriptome specimens, which coalesced on induction of MYC oncogene and Cyclin E/CDK2 activity. We propose that targeting the G1 kinases CDK2, CDK4, and CDK6 with a small-molecule overcomes resistance to CDK4/6 inhibition. We describe the pharmacodynamics and efficacy of PF-06873600 (PF3600), a pyridopyrimidine with potent inhibition of CDK2/4/6 activity and efficacy in multiple in vivo tumor models. Together with the clinical analysis, MYC activity predicts (PF3600) efficacy across multiple cell lineages. Finally, we find that CDK2/4/6 inhibition does not compromise tumor-specific immune checkpoint blockade responses in syngeneic models. We anticipate that (PF3600), currently in phase 1 clinical trials, offers a therapeutic option to cancer patients in whom CDK4/6 inhibition is insufficient to alter disease progression.
Collapse
Affiliation(s)
- Kevin Freeman-Cook
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Robert L Hoffman
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Nichol Miller
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Jonathan Almaden
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - John Chionis
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Qin Zhang
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Koleen Eisele
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Chaoting Liu
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Cathy Zhang
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Nanni Huser
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Lisa Nguyen
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Cinthia Costa-Jones
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Sherry Niessen
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Jordan Carelli
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - John Lapek
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Scott L Weinrich
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Ping Wei
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Elizabeth McMillan
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Elizabeth Wilson
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Tim S Wang
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Michele McTigue
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Rose Ann Ferre
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - You-Ai He
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Sacha Ninkovic
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Douglas Behenna
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Khanh T Tran
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Scott Sutton
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Asako Nagata
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Martha A Ornelas
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Susan E Kephart
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Luke R Zehnder
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Brion Murray
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Meirong Xu
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - James E Solowiej
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Ravi Visswanathan
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Britton Boras
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - David Looper
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Nathan Lee
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Jadwiga R Bienkowska
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Zhou Zhu
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Zhengyan Kan
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Ying Ding
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Xinmeng Jasmine Mu
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Cecilia Oderup
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Shahram Salek-Ardakani
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Michael A White
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Todd VanArsdale
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA.
| | - Stephen G Dann
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA.
| |
Collapse
|
10
|
Terse A, Amin N, Hall B, Bhaskar M, Binukumar B, Utreras E, Pareek TK, Pant H, Kulkarni AB. Protocols for Characterization of Cdk5 Kinase Activity. Curr Protoc 2021; 1:e276. [PMID: 34679246 PMCID: PMC8555461 DOI: 10.1002/cpz1.276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cyclin-dependent kinases (Cdks) are generally known to be involved in controlling the cell cycle, but Cdk5 is a unique member of this protein family for being most active in post-mitotic neurons. Cdk5 is developmentally important in regulating neuronal migration, neurite outgrowth, and axon guidance. Cdk5 is enriched in synaptic membranes and is known to modulate synaptic activity. Postnatally, Cdk5 can also affect neuronal processes such as dopaminergic signaling and pain sensitivity. Dysregulated Cdk5, in contrast, has been linked to neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Despite primarily being implicated in neuronal development and activity, Cdk5 has lately been linked to non-neuronal functions including cancer cell growth, immune responses, and diabetes. Since Cdk5 activity is tightly regulated, a method for measuring its kinase activity is needed to fully understand the precise role of Cdk5 in developmental and disease processes. This article includes methods for detecting Cdk5 kinase activity in cultured cells or tissues, identifying new substrates, and screening for new kinase inhibitors. Furthermore, since Cdk5 shares homology and substrate specificity with Cdk1 and Cdk2, the Cdk5 kinase assay can be used, with modification, to measure the activity of other Cdks as well. © 2021 Wiley Periodicals LLC. This article has been contributed to by US Government employees and their work is in the public domain in the USA. Basic Protocol 1: Measuring Cdk5 activity from protein lysates Support Protocol 1: Immunoprecipitation of Cdk5 using Dynabeads Alternate Protocol: Non-radioactive protocols to measure Cdk5 kinase activity Support Protocol 2: Western blot analysis for the detection of Cdk5, p35, and p39 Support Protocol 3: Immunodetection analysis for Cdk5, p35, and p39 Support Protocol 4: Genetically engineered mice (+ and - controls) Basic Protocol 2: Identifying new Cdk5 substrates and kinase inhibitors.
Collapse
Affiliation(s)
- Anita Terse
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Niranjana Amin
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Bradford Hall
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Manju Bhaskar
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - B.K Binukumar
- CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
| | - Elias Utreras
- Department of Biology, Universidad de Chile, Santiago, Chile
| | | | - Harish Pant
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Ashok B. Kulkarni
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
11
|
Dobyns WB. The Names of Things: The 2018 Bernard Sachs Lecture. Pediatr Neurol 2021; 122:41-49. [PMID: 34330614 DOI: 10.1016/j.pediatrneurol.2021.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 11/22/2022]
Abstract
In 2018, I was honored to receive the Bernard Sachs Award for a lifetime of work expanding knowledge of diverse neurodevelopmental disorders. Summarizing work over more than 30 years is difficult but is an opportunity to chronicle the dramatic changes in the medical and scientific world that have transformed the field of Child Neurology over this time, as reflected in my own work. Here I have chosen to highlight five broad themes of my research beginning with my interest in descriptive terms that drive wider understanding and my choice for the title of this review. From there I will go on to contrast the state of knowledge as I entered the field with the state of knowledge today for four human brain malformations-lissencephaly, megalencephaly, cerebellar malformations, and polymicrogyria. For all, the changes have been dramatic.
Collapse
Affiliation(s)
- William B Dobyns
- Division of Genetics and Metabolism, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
12
|
Hickmott RA, Bosakhar A, Quezada S, Barresi M, Walker DW, Ryan AL, Quigley A, Tolcos M. The One-Stop Gyrification Station - Challenges and New Technologies. Prog Neurobiol 2021; 204:102111. [PMID: 34166774 DOI: 10.1016/j.pneurobio.2021.102111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/31/2021] [Accepted: 06/18/2021] [Indexed: 12/12/2022]
Abstract
The evolution of the folded cortical surface is an iconic feature of the human brain shared by a subset of mammals and considered pivotal for the emergence of higher-order cognitive functions. While our understanding of the neurodevelopmental processes involved in corticogenesis has greatly advanced over the past 70 years of brain research, the fundamental mechanisms that result in gyrification, along with its originating cytoarchitectural location, remain largely unknown. This review brings together numerous approaches to this basic neurodevelopmental problem, constructing a narrative of how various models, techniques and tools have been applied to the study of gyrification thus far. After a brief discussion of core concepts and challenges within the field, we provide an analysis of the significant discoveries derived from the parallel use of model organisms such as the mouse, ferret, sheep and non-human primates, particularly with regard to how they have shaped our understanding of cortical folding. We then focus on the latest developments in the field and the complementary application of newly emerging technologies, such as cerebral organoids, advanced neuroimaging techniques, and atomic force microscopy. Particular emphasis is placed upon the use of novel computational and physical models in regard to the interplay of biological and physical forces in cortical folding.
Collapse
Affiliation(s)
- Ryan A Hickmott
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, 3083, Australia; BioFab3D@ACMD, St Vincent's Hospital Melbourne, Fitzroy, VIC, 3065, Australia
| | - Abdulhameed Bosakhar
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, 3083, Australia
| | - Sebastian Quezada
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, 3083, Australia
| | - Mikaela Barresi
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, 3083, Australia
| | - David W Walker
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, 3083, Australia
| | - Amy L Ryan
- Hastings Centre for Pulmonary Research, Department of Pulmonary, Critical Care and Sleep Medicine, USC Keck School of Medicine, University of Southern California, CA, USA and Department of Stem Cell and Regenerative Medicine, University of Southern California, CA, 90033, USA
| | - Anita Quigley
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, 3083, Australia; BioFab3D@ACMD, St Vincent's Hospital Melbourne, Fitzroy, VIC, 3065, Australia; School of Engineering, RMIT University, Melbourne, VIC, 3000, Australia; Department of Medicine, University of Melbourne, St Vincent's Hospital, Fitzroy, VIC, 3065, Australia; ARC Centre of Excellence in Electromaterials Science, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Mary Tolcos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, 3083, Australia.
| |
Collapse
|
13
|
Kawasaki H. [Investigation of the Mechanisms Underlying Development and Diseases of the Cerebral Cortex Using Mice and Ferrets]. YAKUGAKU ZASSHI 2021; 141:349-357. [PMID: 33642503 DOI: 10.1248/yakushi.20-00198-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Folds of the cerebral cortex, which are called gyri and sulci, are one of the most prominent features of the mammalian brain. However, the mechanisms underlying the development and malformation of cortical folds are largely unknown, mainly because they are difficult to investigate in mice, whose brain do not have cortical folds. To investigate the mechanisms underlying the development and malformation of cortical folds, we developed a genetic manipulation technique for the cerebral cortex of gyrencephalic carnivore ferrets. Genes-of-interest can be expressed in the ferret cortex rapidly and efficiently. We also demonstrated that genes-of-interest can be knocked out in the ferret cortex by combining in utero electroporation and the CRISPR/Cas9 system. Using our technique, we found that fibroblast growth factor (FGF) signaling and sonic hedgehog (Shh) signaling are crucial for cortical folding. In addition, we found that FGF signaling and Shh signaling preferentially increased outer radial glial cells and the thickness of upper layers of the cerebral cortex. Furthermore, over-activation of FGF signaling and Shh signaling resulted in polymicrogyria. Our findings provide in vivo data about the mechanisms of cortical folding in gyrencephalic mammals. Our technique for the ferret cerebral cortex should be useful for investigating the mechanisms underlying the development and diseases of the cerebral cortex that cannot be investigated using mice.
Collapse
Affiliation(s)
- Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medicine, Kanazawa University
| |
Collapse
|
14
|
Doornbos C, Roepman R. Moonlighting of mitotic regulators in cilium disassembly. Cell Mol Life Sci 2021; 78:4955-4972. [PMID: 33860332 PMCID: PMC8233288 DOI: 10.1007/s00018-021-03827-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 03/03/2021] [Accepted: 03/27/2021] [Indexed: 02/07/2023]
Abstract
Correct timing of cellular processes is essential during embryological development and to maintain the balance between healthy proliferation and tumour formation. Assembly and disassembly of the primary cilium, the cell’s sensory signalling organelle, are linked to cell cycle timing in the same manner as spindle pole assembly and chromosome segregation. Mitotic processes, ciliary assembly, and ciliary disassembly depend on the centrioles as microtubule-organizing centres (MTOC) to regulate polymerizing and depolymerizing microtubules. Subsequently, other functional protein modules are gathered to potentiate specific protein–protein interactions. In this review, we show that a significant subset of key mitotic regulator proteins is moonlighting at the cilium, among which PLK1, AURKA, CDC20, and their regulators. Although ciliary assembly defects are linked to a variety of ciliopathies, ciliary disassembly defects are more often linked to brain development and tumour formation. Acquiring a better understanding of the overlap in regulators of ciliary disassembly and mitosis is essential in finding therapeutic targets for the different diseases and types of tumours associated with these regulators.
Collapse
Affiliation(s)
- Cenna Doornbos
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.,Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ronald Roepman
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands. .,Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
15
|
Sasabayashi D, Takahashi T, Takayanagi Y, Suzuki M. Anomalous brain gyrification patterns in major psychiatric disorders: a systematic review and transdiagnostic integration. Transl Psychiatry 2021; 11:176. [PMID: 33731700 PMCID: PMC7969935 DOI: 10.1038/s41398-021-01297-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 02/14/2021] [Accepted: 02/24/2021] [Indexed: 01/31/2023] Open
Abstract
Anomalous patterns of brain gyrification have been reported in major psychiatric disorders, presumably reflecting their neurodevelopmental pathology. However, previous reports presented conflicting results of patients having hyper-, hypo-, or normal gyrification patterns and lacking in transdiagnostic consideration. In this article, we systematically review previous magnetic resonance imaging studies of brain gyrification in schizophrenia, bipolar disorder, major depressive disorder, and autism spectrum disorder at varying illness stages, highlighting the gyral pattern trajectory for each disorder. Patients with each psychiatric disorder may exhibit deviated primary gyri formation under neurodevelopmental genetic control in their fetal life and infancy, and then exhibit higher-order gyral changes due to mechanical stress from active brain changes (e.g., progressive reduction of gray matter volume and white matter integrity) thereafter, representing diversely altered pattern trajectories from those of healthy controls. Based on the patterns of local connectivity and changes in neurodevelopmental gene expression in major psychiatric disorders, we propose an overarching model that spans the diagnoses to explain how deviated gyral pattern trajectories map onto clinical manifestations (e.g., psychosis, mood dysregulation, and cognitive impairments), focusing on the common and distinct gyral pattern changes across the disorders in addition to their correlations with specific clinical features. This comprehensive understanding of the role of brain gyrification pattern on the pathophysiology may help to optimize the prediction and diagnosis of psychiatric disorders using objective biomarkers, as well as provide a novel nosology informed by neural circuits beyond the current descriptive diagnostics.
Collapse
Affiliation(s)
- Daiki Sasabayashi
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama, Japan. .,Research Center for Idling Brain Science, University of Toyama, Toyama, Japan.
| | - Tsutomu Takahashi
- grid.267346.20000 0001 2171 836XDepartment of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama, Japan ,grid.267346.20000 0001 2171 836XResearch Center for Idling Brain Science, University of Toyama, Toyama, Japan
| | - Yoichiro Takayanagi
- grid.267346.20000 0001 2171 836XDepartment of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama, Japan ,Arisawabashi Hospital, Toyama, Japan
| | - Michio Suzuki
- grid.267346.20000 0001 2171 836XDepartment of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama, Japan ,grid.267346.20000 0001 2171 836XResearch Center for Idling Brain Science, University of Toyama, Toyama, Japan
| |
Collapse
|
16
|
Łukasik P, Załuski M, Gutowska I. Cyclin-Dependent Kinases (CDK) and Their Role in Diseases Development-Review. Int J Mol Sci 2021; 22:ijms22062935. [PMID: 33805800 PMCID: PMC7998717 DOI: 10.3390/ijms22062935] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/07/2021] [Accepted: 03/09/2021] [Indexed: 12/13/2022] Open
Abstract
Cyclin-dependent kinases (CDKs) are involved in many crucial processes, such as cell cycle and transcription, as well as communication, metabolism, and apoptosis. The kinases are organized in a pathway to ensure that, during cell division, each cell accurately replicates its DNA, and ensure its segregation equally between the two daughter cells. Deregulation of any of the stages of the cell cycle or transcription leads to apoptosis but, if uncorrected, can result in a series of diseases, such as cancer, neurodegenerative diseases (Alzheimer’s or Parkinson’s disease), and stroke. This review presents the current state of knowledge about the characteristics of cyclin-dependent kinases as potential pharmacological targets.
Collapse
Affiliation(s)
- Paweł Łukasik
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstancow Wlkp. 72 Av., 70-111 Szczecin, Poland;
| | - Michał Załuski
- Department of Pharmaceutical Chemistry, Pomeranian Medical University in Szczecin, Powstancow Wlkp. 72 Av., 70-111 Szczecin, Poland;
| | - Izabela Gutowska
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstancow Wlkp. 72 Av., 70-111 Szczecin, Poland;
- Correspondence:
| |
Collapse
|
17
|
Klingler E, Francis F, Jabaudon D, Cappello S. Mapping the molecular and cellular complexity of cortical malformations. Science 2021; 371:371/6527/eaba4517. [PMID: 33479124 DOI: 10.1126/science.aba4517] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The cerebral cortex is an intricate structure that controls human features such as language and cognition. Cortical functions rely on specialized neurons that emerge during development from complex molecular and cellular interactions. Neurodevelopmental disorders occur when one or several of these steps is incorrectly executed. Although a number of causal genes and disease phenotypes have been identified, the sequence of events linking molecular disruption to clinical expression mostly remains obscure. Here, focusing on human malformations of cortical development, we illustrate how complex interactions at the genetic, cellular, and circuit levels together contribute to diversity and variability in disease phenotypes. Using specific examples and an online resource, we propose that a multilevel assessment of disease processes is key to identifying points of vulnerability and developing new therapeutic strategies.
Collapse
Affiliation(s)
- Esther Klingler
- Department of Basic Neurosciences, University of Geneva, CH-1202 Geneva, Switzerland
| | - Fiona Francis
- INSERM U 1270, F-75005 Paris, France.,Sorbonne University, UMR-S 1270, F-75005 Paris, France.,Institut du Fer à Moulin, F-75005 Paris, France
| | - Denis Jabaudon
- Department of Basic Neurosciences, University of Geneva, CH-1202 Geneva, Switzerland. .,Clinic of Neurology, Geneva University Hospital, 1211 Geneva, Switzerland
| | | |
Collapse
|
18
|
Franchini LF. Genetic Mechanisms Underlying Cortical Evolution in Mammals. Front Cell Dev Biol 2021; 9:591017. [PMID: 33659245 PMCID: PMC7917222 DOI: 10.3389/fcell.2021.591017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
The remarkable sensory, motor, and cognitive abilities of mammals mainly depend on the neocortex. Thus, the emergence of the six-layered neocortex in reptilian ancestors of mammals constitutes a fundamental evolutionary landmark. The mammalian cortex is a columnar epithelium of densely packed cells organized in layers where neurons are generated mainly in the subventricular zone in successive waves throughout development. Newborn cells move away from their site of neurogenesis through radial or tangential migration to reach their specific destination closer to the pial surface of the same or different cortical area. Interestingly, the genetic programs underlying neocortical development diversified in different mammalian lineages. In this work, I will review several recent studies that characterized how distinct transcriptional programs relate to the development and functional organization of the neocortex across diverse mammalian lineages. In some primates such as the anthropoids, the neocortex became extremely large, especially in humans where it comprises around 80% of the brain. It has been hypothesized that the massive expansion of the cortical surface and elaboration of its connections in the human lineage, has enabled our unique cognitive capacities including abstract thinking, long-term planning, verbal language and elaborated tool making capabilities. I will also analyze the lineage-specific genetic changes that could have led to the modification of key neurodevelopmental events, including regulation of cell number, neuronal migration, and differentiation into specific phenotypes, in order to shed light on the evolutionary mechanisms underlying the diversity of mammalian brains including the human brain.
Collapse
Affiliation(s)
- Lucía Florencia Franchini
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
19
|
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
|
20
|
Kolbjer S, Martin DA, Pettersson M, Dahlin M, Anderlid BM. Lissencephaly in an epilepsy cohort: Molecular, radiological and clinical aspects. Eur J Paediatr Neurol 2021; 30:71-81. [PMID: 33453472 DOI: 10.1016/j.ejpn.2020.12.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 12/22/2020] [Accepted: 12/27/2020] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Lissencephaly is a rare malformation of cortical development due to abnormal transmantle migration resulting in absent or reduced gyration. The lissencephaly spectrum consists of agyria, pachygyria and subcortical band heterotopia. In this study we compared genetic aetiology, neuroradiology, clinical phenotype and response to antiepileptic drugs in patients with epilepsy and lissencephaly spectrum malformations. METHODS The study group consisted of 20 patients - 13 males and 7 females, aged 18 months to 21 years at the time of data collection. Genetic testing was performed by oligonucleotide array comparative genomic hybridization (microarray), multiplex ligation-dependent probe amplification (MLPA), targeted gene panels and whole exome/genome sequencing. All neuroradiological investigations were re-evaluated and the malformations were classified by the same neuroradiologist. Clinical features and response to anti-epileptic drugs (AEDs) were evaluated by retrospective review of medical records. RESULTS In eleven patients (55%) mutations in PAFAH1B1 (LIS1) or variable microdeletions of 17p13.3 including the PAFAH1B1 gene were detected. Four patients (20%) had tubulin encoding gene mutations (TUBA1A, TUBG1 and TUBGCP6). Mutations in DCX, DYNC1H1, ADGRG1 and WDR62 were identified in single patients. In one patient, a possibly pathogenic intragenic deletion in TRIO was detected. A clear radiologic distinction could be made between tubulinopathies and PAFAH1B1 related lissencephaly. The majority of the patients had therapy resistant epilepsy and epileptic spasms was the most prominent seizure type. The best therapeutic response to seizure control in our cohort was obtained by the ketogenic diet, vigabatrin, clobazam, phenobarbital and valproate. CONCLUSION The most common genetic aetiologies in our cohort of 20 individuals with epilepsy and lissencephaly spectrum were intragenic deletions or single nucleotide mutations in PAFAH1B1 or larger deletions in 17p13.3, encompassing PAFAH1B1, followed by mutations in tubulin encoding genes. Radiological findings could reliably predict molecular results only in agyria with a posterior to anterior gradient. Radiological and molecular findings did not correlate consistently with severity of clinical outcome or therapeutic response.
Collapse
Affiliation(s)
- Sintia Kolbjer
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Neuropaediatrics, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden.
| | - Daniel A Martin
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden; Department of Paediatric Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Pettersson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Dahlin
- Department of Neuropaediatrics, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden; Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Britt-Marie Anderlid
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
21
|
Fourel G, Boscheron C. Tubulin mutations in neurodevelopmental disorders as a tool to decipher microtubule function. FEBS Lett 2020; 594:3409-3438. [PMID: 33064843 DOI: 10.1002/1873-3468.13958] [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: 12/13/2019] [Revised: 09/28/2020] [Accepted: 10/05/2020] [Indexed: 01/08/2023]
Abstract
Malformations of cortical development (MCDs) are a group of severe brain malformations associated with intellectual disability and refractory childhood epilepsy. Human missense heterozygous mutations in the 9 α-tubulin and 10 β-tubulin isoforms forming the heterodimers that assemble into microtubules (MTs) were found to cause MCDs. However, how a single mutated residue in a given tubulin isoform can perturb the entire microtubule population in a neuronal cell remains a crucial question. Here, we examined 85 MCD-associated tubulin mutations occurring in TUBA1A, TUBB2, and TUBB3 and their location in a three-dimensional (3D) microtubule cylinder. Mutations hitting residues exposed on the outer microtubule surface are likely to alter microtubule association with partners, while alteration of intradimer contacts may impair dimer stability and straightness. Other types of mutations are predicted to alter interdimer and lateral contacts, which are responsible for microtubule cohesion, rigidity, and dynamics. MCD-associated tubulin mutations surprisingly fall into all categories, thus providing unexpected insights into how a single mutation may impair microtubule function and elicit dominant effects in neurons.
Collapse
|
22
|
Colas P. Cyclin-dependent kinases and rare developmental disorders. Orphanet J Rare Dis 2020; 15:203. [PMID: 32762766 PMCID: PMC7410148 DOI: 10.1186/s13023-020-01472-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022] Open
Abstract
Extensive studies in the past 30 years have established that cyclin-dependent kinases (CDKs) exert many diverse, important functions in a number of molecular and cellular processes that are at play during development. Not surprisingly, mutations affecting CDKs or their activating cyclin subunits have been involved in a variety of rare human developmental disorders. These recent findings are reviewed herein, giving a particular attention to the discovered mutations and their demonstrated or hypothesized functional consequences, which can account for pathological human phenotypes. The review highlights novel, important CDK or cyclin functions that were unveiled by their association with human disorders, and it discusses the shortcomings of mouse models to reveal some of these functions. It explains how human genetics can be used in combination with proteome-scale interaction databases to loom regulatory networks around CDKs and cyclins. Finally, it advocates the use of these networks to profile pathogenic CDK or cyclin variants, in order to gain knowledge on protein function and on pathogenic mechanisms.
Collapse
Affiliation(s)
- Pierre Colas
- Laboratory of Integrative Biology of Marine Models, Station Biologique de Roscoff, Sorbonne Université / CNRS, Roscoff, France.
| |
Collapse
|
23
|
Quezada S, van de Looij Y, Hale N, Rana S, Sizonenko SV, Gilchrist C, Castillo-Melendez M, Tolcos M, Walker DW. Genetic and microstructural differences in the cortical plate of gyri and sulci during gyrification in fetal sheep. Cereb Cortex 2020; 30:6169-6190. [PMID: 32609332 DOI: 10.1093/cercor/bhaa171] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 05/29/2020] [Accepted: 05/29/2020] [Indexed: 12/28/2022] Open
Abstract
Gyrification of the cerebral cortex is a developmentally important process, but the mechanisms that drive cortical folding are not fully known. Theories propose that changes within the cortical plate (CP) cause gyrification, yet differences between the CP below gyri and sulci have not been investigated. Here we report genetic and microstructural differences in the CP below gyri and sulci assessed before (at 70 days of gestational age [GA] 70), during (GA 90), and after (GA 110) gyrification in fetal sheep. The areal density of BDNF, CDK5, and NeuroD6 immunopositive cells were increased, and HDAC5 and MeCP2 mRNA levels were decreased in the CP below gyri compared with sulci during gyrification, but not before. Only the areal density of BDNF-immunopositive cells remained increased after gyrification. MAP2 immunoreactivity and neurite outgrowth were also increased in the CP below gyri compared with sulci at GA 90, and this was associated with microstructural changes assessed via diffusion tensor imaging and neurite orientation dispersion and density imaging at GA 98. Differential neurite outgrowth may therefore explain the localized changes in CP architecture that result in gyrification.
Collapse
Affiliation(s)
- Sebastian Quezada
- The Ritchie Centre, Hudson Institute of Medical Research, Monash University, Clayton, VIC 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia.,School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083 Australia
| | - Yohan van de Looij
- Division of Development and Growth, Department of Paediatrics and Gynaecology-Obstetrics, School of Medicine, University of Geneva, 1204 Geneva, Switzerland.,Functional and Metabolic Imaging Lab, Federal Institute of Technology of Lausanne, Lausanne 1015, Switzerland
| | - Nadia Hale
- The Ritchie Centre, Hudson Institute of Medical Research, Monash University, Clayton, VIC 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Shreya Rana
- The Ritchie Centre, Hudson Institute of Medical Research, Monash University, Clayton, VIC 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Stéphane V Sizonenko
- Division of Development and Growth, Department of Paediatrics and Gynaecology-Obstetrics, School of Medicine, University of Geneva, 1204 Geneva, Switzerland
| | - Courtney Gilchrist
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083 Australia.,Clinical Sciences, Murdoch Children's Research Institute, Parkville, VIC 3052, Australia
| | - Margie Castillo-Melendez
- The Ritchie Centre, Hudson Institute of Medical Research, Monash University, Clayton, VIC 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Mary Tolcos
- The Ritchie Centre, Hudson Institute of Medical Research, Monash University, Clayton, VIC 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia.,School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083 Australia
| | - David W Walker
- The Ritchie Centre, Hudson Institute of Medical Research, Monash University, Clayton, VIC 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia.,School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083 Australia
| |
Collapse
|
24
|
Varderidou-Minasian S, Hinz L, Hagemans D, Posthuma D, Altelaar M, Heine VM. Quantitative proteomic analysis of Rett iPSC-derived neuronal progenitors. Mol Autism 2020; 11:38. [PMID: 32460858 PMCID: PMC7251722 DOI: 10.1186/s13229-020-00344-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 05/04/2020] [Indexed: 12/12/2022] Open
Abstract
Background Rett syndrome (RTT) is a progressive neurodevelopmental disease that is characterized by abnormalities in cognitive, social, and motor skills. RTT is often caused by mutations in the X-linked gene encoding methyl-CpG binding protein 2 (MeCP2). The mechanism by which impaired MeCP2 induces the pathological abnormalities in the brain is not understood. Both patients and mouse models have shown abnormalities at molecular and cellular level before typical RTT-associated symptoms appear. This implies that underlying mechanisms are already affected during neurodevelopmental stages. Methods To understand the molecular mechanisms involved in disease onset, we used an RTT patient induced pluripotent stem cell (iPSC)-based model with isogenic controls and performed time-series of proteomic analysis using in-depth high-resolution quantitative mass spectrometry during early stages of neuronal development. Results We provide mass spectrometry-based quantitative proteomic data, depth of about 7000 proteins, at neuronal progenitor developmental stages of RTT patient cells and isogenic controls. Our data gives evidence of proteomic alteration at early neurodevelopmental stages, suggesting alterations long before the phase that symptoms of RTT syndrome become apparent. Significant changes are associated with the GO enrichment analysis in biological processes cell-cell adhesion, actin cytoskeleton organization, neuronal stem cell population maintenance, and pituitary gland development, next to protein changes previously associated with RTT, i.e., dendrite morphology and synaptic deficits. Differential expression increased from early to late neural stem cell phases, although proteins involved in immunity, metabolic processes, and calcium signaling were affected throughout all stages analyzed. Limitations The limitation of our study is the number of RTT patients analyzed. As the aim of our study was to investigate a large number of proteins, only one patient was considered, of which 3 different RTT iPSC clones and 3 isogenic control iPSC clones were included. Even though this approach allowed the study of mutation-induced alterations due to the usage of isogenic controls, results should be validated on different RTT patients to suggest common disease mechanisms. Conclusions During early neuronal differentiation, there are consistent and time-point specific proteomic alterations in RTT patient cells carrying exons 3–4 deletion in MECP2. We found changes in proteins involved in pathway associated with RTT phenotypes, including dendrite morphology and synaptogenesis. Our results provide a valuable resource of proteins and pathways for follow-up studies, investigating common mechanisms involved during early disease stages of RTT syndrome.
Collapse
Affiliation(s)
- Suzy Varderidou-Minasian
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, 3584, CH, Utrecht, The Netherlands.,Netherlands Proteomics Center, Padualaan 8, 3584, CH, Utrecht, The Netherlands
| | - Lisa Hinz
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Dominique Hagemans
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, 3584, CH, Utrecht, The Netherlands.,Netherlands Proteomics Center, Padualaan 8, 3584, CH, Utrecht, The Netherlands
| | - Danielle Posthuma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Child and Youth Psychiatry, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, 3584, CH, Utrecht, The Netherlands.,Netherlands Proteomics Center, Padualaan 8, 3584, CH, Utrecht, The Netherlands
| | - Vivi M Heine
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands. .,Child and Youth Psychiatry, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
25
|
Kodani A, Kenny C, Lai A, Gonzalez DM, Stronge E, Sejourne GM, Isacco L, Partlow JN, O'Donnell A, McWalter K, Byrne AB, Barkovich AJ, Yang E, Hill RS, Gawlinski P, Wiszniewski W, Cohen JS, Fatemi SA, Baranano KW, Sahin M, Vossler DG, Yuskaitis CJ, Walsh CA. Posterior Neocortex-Specific Regulation of Neuronal Migration by CEP85L Identifies Maternal Centriole-Dependent Activation of CDK5. Neuron 2020; 106:246-255.e6. [PMID: 32097629 PMCID: PMC7255387 DOI: 10.1016/j.neuron.2020.01.030] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/22/2019] [Accepted: 01/22/2020] [Indexed: 01/01/2023]
Abstract
Genes mutated in human neuronal migration disorders encode tubulin proteins and a variety of tubulin-binding and -regulating proteins, but it is very poorly understood how these proteins function together to coordinate migration. Additionally, the way in which regional differences in neocortical migration are controlled is completely unknown. Here we describe a new syndrome with remarkably region-specific effects on neuronal migration in the posterior cortex, reflecting de novo variants in CEP85L. We show that CEP85L is required cell autonomously in vivo and in vitro for migration, that it localizes to the maternal centriole, and that it forms a complex with many other proteins required for migration, including CDK5, LIS1, NDE1, KIF2A, and DYNC1H1. Loss of CEP85L disrupts CDK5 localization and activation, leading to centrosome disorganization and disrupted microtubule cytoskeleton organization. Together, our findings suggest that CEP85L highlights a complex that controls CDK5 activity to promote neuronal migration.
Collapse
Affiliation(s)
- Andrew Kodani
- Division of Genetics and Genomics and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA.
| | - Connor Kenny
- Division of Genetics and Genomics and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA
| | - Abbe Lai
- Division of Genetics and Genomics and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA; Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Dilenny M Gonzalez
- Division of Genetics and Genomics and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA
| | - Edward Stronge
- Division of Genetics and Genomics and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA
| | - Gabrielle M Sejourne
- Division of Genetics and Genomics and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA
| | - Laura Isacco
- Division of Genetics and Genomics and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA
| | - Jennifer N Partlow
- Division of Genetics and Genomics and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA
| | - Anne O'Donnell
- Division of Genetics and Genomics and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA; Center for Mendelian Genomics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Alicia B Byrne
- Center for Mendelian Genomics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Genetics and Molecular Pathology, Centre for Cancer Biology, SA Pathology, Adelaide, SA, Australia; School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - A James Barkovich
- Department of Radiology, University of California, San Francisco, San Francisco, CA, USA
| | - Edward Yang
- Department of Radiology, Boston Children's Hospital, Boston, MA, USA
| | - R Sean Hill
- Division of Genetics and Genomics and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA
| | - Pawel Gawlinski
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland
| | - Wojciech Wiszniewski
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland; Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Julie S Cohen
- Division of Neurogenetics, Kennedy Krieger Institute, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - S Ali Fatemi
- Division of Neurogenetics, Kennedy Krieger Institute, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kristin W Baranano
- Division of Neurogenetics, Kennedy Krieger Institute, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mustafa Sahin
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - David G Vossler
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Christopher J Yuskaitis
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Division of Epilepsy and Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Christopher A Walsh
- Division of Genetics and Genomics and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
26
|
Ghosh SG, Wang L, Breuss MW, Green JD, Stanley V, Yang X, Ross D, Traynor BJ, Alhashem AM, Azam M, Selim L, Bastaki L, Elbastawisy HI, Temtamy S, Zaki M, Gleeson JG. Recurrent homozygous damaging mutation in TMX2, encoding a protein disulfide isomerase, in four families with microlissencephaly. J Med Genet 2020; 57:274-282. [PMID: 31586943 PMCID: PMC7405652 DOI: 10.1136/jmedgenet-2019-106409] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/26/2019] [Accepted: 09/06/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Protein disulfide isomerase (PDI) proteins are part of the thioredoxin protein superfamily. PDIs are involved in the formation and rearrangement of disulfide bonds between cysteine residues during protein folding in the endoplasmic reticulum and are implicated in stress response pathways. METHODS Eight children from four consanguineous families residing in distinct geographies within the Middle East and Central Asia were recruited for study. All probands showed structurally similar microcephaly with lissencephaly (microlissencephaly) brain malformations. DNA samples from each family underwent whole exome sequencing, assessment for repeat expansions and confirmatory segregation analysis. RESULTS An identical homozygous variant in TMX2 (c.500G>A), encoding thioredoxin-related transmembrane protein 2, segregated with disease in all four families. This variant changed the last coding base of exon 6, and impacted mRNA stability. All patients presented with microlissencephaly, global developmental delay, intellectual disability and epilepsy. While TMX2 is an activator of cellular C9ORF72 repeat expansion toxicity, patients showed no evidence of C9ORF72 repeat expansions. CONCLUSION The TMX2 c.500G>A allele associates with recessive microlissencephaly, and patients show no evidence of C9ORF72 expansions. TMX2 is the first PDI implicated in a recessive disease, suggesting a protein isomerisation defect in microlissencephaly.
Collapse
Affiliation(s)
- Shereen Georges Ghosh
- Neurosciences, University of California San Diego, La Jolla, California, USA
- Rady Children's Institute for Genomic Medicine, San Diego, California, USA
| | - Lu Wang
- Neurosciences, University of California San Diego, La Jolla, California, USA
- Rady Children's Institute for Genomic Medicine, San Diego, California, USA
| | - Martin W Breuss
- Neurosciences, University of California San Diego, La Jolla, California, USA
- Rady Children's Institute for Genomic Medicine, San Diego, California, USA
| | - Joshua D Green
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institutes of Health, Bethesda, Maryland, USA
| | - Valentina Stanley
- Neurosciences, University of California San Diego, La Jolla, California, USA
- Rady Children's Institute for Genomic Medicine, San Diego, California, USA
| | - Xiaoxu Yang
- Neurosciences, University of California San Diego, La Jolla, California, USA
- Rady Children's Institute for Genomic Medicine, San Diego, California, USA
| | - Danica Ross
- Neurosciences, University of California San Diego, La Jolla, California, USA
- Rady Children's Institute for Genomic Medicine, San Diego, California, USA
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institutes of Health, Bethesda, Maryland, USA
- Neurology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Amal M Alhashem
- Pediatrics, Prince Sultan Military Medical City, Riyadh, Al Riyadh, Saudi Arabia
| | - Matloob Azam
- Pediatrics and Child Neurology, Wah Medical College, Wah Cantt, Pakistan
| | - Laila Selim
- Pediatric Neurology, Cairo University, Cairo, Egypt
| | - Laila Bastaki
- Kuwait Medical Genetics Centre, Maternity Hospital, Shuwaikh, Kuwait
| | - Hanan I Elbastawisy
- Ophthalmic Genetics, Research Institute of Ophthalmology, Sulaibikhat, Egypt
| | - Samia Temtamy
- Center of Excellence for Human Genetics, National Research Centre, Cairo, Egypt
| | - Maha Zaki
- Clinical Genetics, National Research Centre, Cairo, Egypt
| | - Joseph G Gleeson
- Rady Children's Institute for Genomic Medicine, San Diego, California, USA
- Department of Neuroscience and Pediatrics, Howard Hughes Medical Institute, La Jolla, California, USA
| |
Collapse
|
27
|
Accogli A, Addour-Boudrahem N, Srour M. Neurogenesis, neuronal migration, and axon guidance. HANDBOOK OF CLINICAL NEUROLOGY 2020; 173:25-42. [PMID: 32958178 DOI: 10.1016/b978-0-444-64150-2.00004-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Development of the central nervous system (CNS) is a complex, dynamic process that involves a precisely orchestrated sequence of genetic, environmental, biochemical, and physical factors from early embryonic stages to postnatal life. Duringthe past decade, great strides have been made to unravel mechanisms underlying human CNS development through the employment of modern genetic techniques and experimental approaches. In this chapter, we review the current knowledge regarding the main developmental processes and signaling mechanisms of (i) neurogenesis, (ii) neuronal migration, and (iii) axon guidance. We discuss mechanisms related to neural stem cells proliferation, migration, terminal translocation of neuronal progenitors, and axon guidance and pathfinding. For each section, we also provide a comprehensive overview of the underlying regulatory processes, including transcriptional, posttranscriptional, and epigenetic factors, and a myriad of signaling pathways that are pivotal to determine the fate of neuronal progenitors and newly formed migrating neurons. We further highlight how impairment of this complex regulating system, such as mutations in its core components, may cause cortical malformation, epilepsy, intellectual disability, and autism in humans. A thorough understanding of normal human CNS development is thus crucial to decipher mechanisms responsible for neurodevelopmental disorders and in turn guide the development of effective and targeted therapeutic strategies.
Collapse
Affiliation(s)
- Andrea Accogli
- Unit of Medical Genetics, Istituto Giannina Gaslini Pediatric Hospital, Genova, Italy; Departments of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal-Child Science, Università degli Studi di Genova, Genova, Italy
| | | | - Myriam Srour
- Research Institute, McGill University Health Centre, Montreal, QC, Canada; Department of Pediatrics, Division of Pediatric Neurology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
28
|
Lee DK, Lee H, Yoon J, Hong S, Lee Y, Kim KT, Kim JW, Song MR. Cdk5 regulates N-cadherin-dependent neuronal migration during cortical development. Biochem Biophys Res Commun 2019; 514:645-652. [PMID: 31076103 DOI: 10.1016/j.bbrc.2019.04.166] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 04/24/2019] [Indexed: 02/06/2023]
Abstract
Cyclin-dependent kinase 5 (Cdk5) controls neuronal migration in the developing cortex when multipolar newborn neurons transform to become bipolar. However, by which mechanisms Cdk5 controls cell adhesion in migrating neurons are not fully understood. In this study, we examined the functional interaction between Cdk5 and N-cadherin (Ncad) in newborn neurons when they undergo the multipolar to bipolar transition in the intermediate zone (IZ). Detailed expression analysis revealed that both Cdk5 and Ncad were present in GFP-electroporated migrating neurons in the IZ. Misexpression of dominant negative Cdk5 into the embryonic brains stalled neuronal locomotion in the lower IZ in which arrested cells were round or multipolar. When Ncad was co-introduced with Cdk5DN, however, cells continue to migrate into the cortical plate (CP) and migrating neurons acquired typical bipolar morphology with a pia-directed leading process. Similarly, downregulation of CDK5 resulted in lesser aggregation ability, reversed by the expression of Ncad in vitro. Down-regulation of activity or protein level of CDK5 did not alter the total amount of NCAD proteins but lowered its surface expression in cells. Lastly, expression of CDK5 and NCAD overlapped in the IZ of the human fetal cortex, indicating that the role of Cdk5 and Ncad in neuronal migration is evolutionarily conserved.
Collapse
Affiliation(s)
- Dong-Keun Lee
- School of Life Sciences, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju, 500-712, Republic of Korea
| | - Hojae Lee
- School of Life Sciences, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju, 500-712, Republic of Korea
| | - Jiyoung Yoon
- School of Life Sciences, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju, 500-712, Republic of Korea
| | - Sujeong Hong
- School of Life Sciences, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju, 500-712, Republic of Korea
| | - Yunjeong Lee
- School of Life Sciences, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju, 500-712, Republic of Korea
| | - Kyung-Tai Kim
- Jeonbuk Department of Inhalation Research, Korea Institute of Toxicology, 30 Baekhak1-gil, Jeongeup, Jeollabuk-do, 56212, Republic of Korea
| | - Jong Woon Kim
- Department of Obstetrics and Gynecology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Mi-Ryoung Song
- School of Life Sciences, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju, 500-712, Republic of Korea.
| |
Collapse
|
29
|
Farahani R, Rezaei-Lotfi S, Simonian M, Hunter N. Bi-modal reprogramming of cell cycle by MiRNA-4673 amplifies human neurogenic capacity. Cell Cycle 2019; 18:848-868. [PMID: 30907228 DOI: 10.1080/15384101.2019.1595873] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Molecular mechanisms that inform heterochronic adaptations of neurogenesis in Homo sapiens remain largely unknown. Here, we uncover a signature in the cell cycle that amplifies the proliferative capacity of human neural progenitors by input from microRNA4673 encoded in Notch-1. The miRNA instructs bimodal reprogramming of the cell cycle, leading to initial synchronization of neural precursors at the G0 phase of the cell cycle followed by accelerated progression through interphase. The key event in G0 synchronization is transient inhibition by miR4673 of cyclin-dependent kinase-18, a member of an ancient family of cyclins that license M-G1 transition. In parallel, autophagic degradation of p53/p21 and transcriptional silencing of XRCC3/BRCA2 relax G1/S cell cycle checkpoint and accelerate interphase by ≈2.8-fold. The resultant reprogrammed cell cycle amplifies the proliferative capacity and delays the differentiation of human neural progenitors.
Collapse
Affiliation(s)
- Ramin Farahani
- a IDR/Westmead Institute for Medical Research , Sydney , NSW , Australia.,b Department of Life Sciences, Faculty of Medicine and Health Sciences , University of Sydney , Sydney , NSW , Australia
| | - Saba Rezaei-Lotfi
- b Department of Life Sciences, Faculty of Medicine and Health Sciences , University of Sydney , Sydney , NSW , Australia
| | - Mary Simonian
- a IDR/Westmead Institute for Medical Research , Sydney , NSW , Australia
| | - Neil Hunter
- a IDR/Westmead Institute for Medical Research , Sydney , NSW , Australia.,b Department of Life Sciences, Faculty of Medicine and Health Sciences , University of Sydney , Sydney , NSW , Australia
| |
Collapse
|
30
|
Abstract
Mutations causing dysfunction of the tubulins and microtubule-associated proteins, otherwise known as tubulinopathies, are a group of recently described entities, that lead to complex brain malformations. An understanding of the fundamental principles of operation of the cytoskeleton and compounds in particular microtubules, actin, and microtubule-associated proteins, can assist in the interpretation of the imaging findings of tubulinopathies. Somewhat consistent morphological imaging patterns have been described in tubulinopathies such as dysmorphic basal ganglia-the hallmark (found in 75% of cases), callosal dysgenesis, cerebellar hypoplasia/dysplasia, and cortical malformations, most notably lissencephaly. Recognizing the common imaging phenotypes present in tubulinopathies can prove invaluable in directing the genetic workup for a patient with brain malformations.
Collapse
|
31
|
Platzer K, Sticht H, Edwards SL, Allen W, Angione KM, Bonati MT, Brasington C, Cho MT, Demmer LA, Falik-Zaccai T, Gamble CN, Hellenbroich Y, Iascone M, Kok F, Mahida S, Mandel H, Marquardt T, McWalter K, Panis B, Pepler A, Pinz H, Ramos L, Shinde DN, Smith-Hicks C, Stegmann APA, Stöbe P, Stumpel CTRM, Wilson C, Lemke JR, Di Donato N, Miller KG, Jamra R. De Novo Variants in MAPK8IP3 Cause Intellectual Disability with Variable Brain Anomalies. Am J Hum Genet 2019; 104:203-212. [PMID: 30612693 DOI: 10.1016/j.ajhg.2018.12.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 12/11/2018] [Indexed: 01/28/2023] Open
Abstract
Using exome sequencing, we have identified de novo variants in MAPK8IP3 in 13 unrelated individuals presenting with an overlapping phenotype of mild to severe intellectual disability. The de novo variants comprise six missense variants, three of which are recurrent, and three truncating variants. Brain anomalies such as perisylvian polymicrogyria, cerebral or cerebellar atrophy, and hypoplasia of the corpus callosum were consistent among individuals harboring recurrent de novo missense variants. MAPK8IP3 has been shown to be involved in the retrograde axonal-transport machinery, but many of its specific functions are yet to be elucidated. Using the CRISPR-Cas9 system to target six conserved amino acid positions in Caenorhabditis elegans, we found that two of the six investigated human alterations led to a significantly elevated density of axonal lysosomes, and five variants were associated with adverse locomotion. Reverse-engineering normalized the observed adverse effects back to wild-type levels. Combining genetic, phenotypic, and functional findings, as well as the significant enrichment of de novo variants in MAPK8IP3 within our total cohort of 27,232 individuals who underwent exome sequencing, we implicate de novo variants in MAPK8IP3 as a cause of a neurodevelopmental disorder with intellectual disability and variable brain anomalies.
Collapse
Affiliation(s)
- Konrad Platzer
- Institute of Human Genetics, University of Leipzig Hospitals and Clinics, Leipzig 04103, Germany.
| | - Heinrich Sticht
- Institute of Biochemistry, Emil-Fischer Center, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Stacey L Edwards
- Genetic Models of Disease Laboratory, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - William Allen
- Department of Genetics, Fullerton Genetics Center, Asheville, NC 28803, USA
| | - Kaitlin M Angione
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Maria T Bonati
- Clinic of Medical Genetics, IRCCS Istituto Auxologico Italiano, Milan 20149, Italy
| | - Campbell Brasington
- Department of Pediatrics, Clinical Genetics, Levine Children's Hospital at Carolina Healthcare System, Charlotte, NC 28203, USA
| | | | - Laurie A Demmer
- Department of Pediatrics, Clinical Genetics, Levine Children's Hospital at Carolina Healthcare System, Charlotte, NC 28203, USA
| | - Tzipora Falik-Zaccai
- Institute of Human Genetics, Galilee Medical Center, Nahariya 22100, Israel; The Azrieli School of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Candace N Gamble
- Department of Pediatrics, University of Texas Health Medical School, Houston, TX 77030, USA
| | - Yorck Hellenbroich
- Institute of Human Genetics, University of Lübeck, Lübeck 23562, Germany
| | - Maria Iascone
- Laboratorio di Genetica Medica, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo 24127, Italy
| | - Fernando Kok
- Mendelics Genomic Analysis, São Paulo 04013-000, Brazil
| | - Sonal Mahida
- Department of Neurology, Kennedy Krieger Institute, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hanna Mandel
- Institute of Human Genetics, Galilee Medical Center, Nahariya 22100, Israel
| | - Thorsten Marquardt
- Department of Pediatrics, University Hospital Münster, Münster 48149, Germany
| | | | - Bianca Panis
- Department of Pediatrics, Zuyderland Medical Center, Heerlen and Sittard 6419, the Netherlands
| | - Alexander Pepler
- CeGaT GmbH and Praxis für Humangenetik Tübingen, Tübingen 72076, Germany
| | - Hailey Pinz
- Division of Medical Genetics, Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, MO 63104, USA
| | - Luiza Ramos
- Mendelics Genomic Analysis, São Paulo 04013-000, Brazil
| | - Deepali N Shinde
- Division of Clinical Genomics, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Constance Smith-Hicks
- Department of Neurology, Kennedy Krieger Institute, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alexander P A Stegmann
- Department of Clinical Genetics and School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht 6229, the Netherlands
| | - Petra Stöbe
- CeGaT GmbH and Praxis für Humangenetik Tübingen, Tübingen 72076, Germany
| | - Constance T R M Stumpel
- Department of Clinical Genetics and School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht 6229, the Netherlands
| | - Carolyn Wilson
- Department of Genetics, Fullerton Genetics Center, Asheville, NC 28803, USA
| | - Johannes R Lemke
- Institute of Human Genetics, University of Leipzig Hospitals and Clinics, Leipzig 04103, Germany
| | - Nataliya Di Donato
- Institute for Clinical Genetics, Carl Gustav Carus Faculty of Medicine, TU Dresden, Dresden 01307, Germany
| | - Kenneth G Miller
- Genetic Models of Disease Laboratory, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Rami Jamra
- Institute of Human Genetics, University of Leipzig Hospitals and Clinics, Leipzig 04103, Germany
| |
Collapse
|
32
|
Abstract
Mutations in CDK13 have recently been identified as a novel cause of syndromic intellectual disability. In this chapter, we review the 44 cases of CDK13-related disorder reported to date, highlighting key clinical pointers to this diagnosis including characteristic craniofacial features, feeding difficulties in infancy, and the presence of structural heart or brain malformations. The spectrum of reported mutations is also described, demonstrating an excess of missense mutations arising in the protein kinase domain. Exploration of genotype-phenotype correlations suggests a trend toward milder phenotypes in patients with mutations predicted to cause haploinsufficiency of CDK13, while missense mutations affecting amino acid residue 842 appear most likely to be associated with structural malformations. The greater phenotypic impact of missense variants is hypothesized to occur due to a dominant-negative mechanism, by which the mutant protein acts to sequester cyclin K in inactive complexes. Functional studies to validate this hypothesis have not yet been carried out, however. Differential diagnosis and recommendations for clinical care of patients with CDK13-related disorder are also described, emphasizing baseline echocardiography, vigilance for feeding and swallowing difficulties, and regular developmental evaluation as key components of care. Finally, future directions for CDK13 research are discussed, including the need to resolve uncertainty regarding pathogenicity of CDK13 haploinsufficiency, and to gather further longitudinal data from large cohorts in order to inform the clinical care of patients with this diagnosis.
Collapse
Affiliation(s)
- Mark James Hamilton
- Nottingham Clinical Genetics Service, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom.
| | - Mohnish Suri
- Nottingham Clinical Genetics Service, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom.
| |
Collapse
|
33
|
Stephen J, Maddirevula S, Nampoothiri S, Burke JD, Herzog M, Shukla A, Steindl K, Eskin A, Patil SJ, Joset P, Lee H, Garrett LJ, Yokoyama T, Balanda N, Bodine SP, Tolman NJ, Zerfas PM, Zheng A, Ramantani G, Girisha KM, Rivas C, Suresh PV, Elkahloun A, Alsaif HS, Wakil SM, Mahmoud L, Ali R, Prochazkova M, Kulkarni AB, Ben-Omran T, Colak D, Morris HD, Rauch A, Martinez-Agosto JA, Nelson SF, Alkuraya FS, Gahl WA, Malicdan MCV, Malicdan MCV. Bi-allelic TMEM94 Truncating Variants Are Associated with Neurodevelopmental Delay, Congenital Heart Defects, and Distinct Facial Dysmorphism. Am J Hum Genet 2018; 103:948-967. [PMID: 30526868 DOI: 10.1016/j.ajhg.2018.11.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 11/02/2018] [Indexed: 02/06/2023] Open
Abstract
Neurodevelopmental disorders (NDD) are genetically and phenotypically heterogeneous conditions due to defects in genes involved in development and function of the nervous system. Individuals with NDD, in addition to their primary neurodevelopmental phenotype, may also have accompanying syndromic features that can be very helpful diagnostically especially those with recognizable facial appearance. In this study, we describe ten similarly affected individuals from six unrelated families of different ethnic origins having bi-allelic truncating variants in TMEM94, which encodes for an uncharacterized transmembrane nuclear protein that is highly conserved across mammals. The affected individuals manifested with global developmental delay/intellectual disability, and dysmorphic facial features including triangular face, deep set eyes, broad nasal root and tip and anteverted nostrils, thick arched eye brows, hypertrichosis, pointed chin, and hypertelorism. Birthweight in the upper normal range was observed in most, and all but one had congenital heart defects (CHD). Gene expression analysis in available cells from affected individuals showed reduced expression of TMEM94. Global transcriptome profiling using microarray and RNA sequencing revealed several dysregulated genes essential for cell growth, proliferation and survival that are predicted to have an impact on cardiotoxicity hematological system and neurodevelopment. Loss of Tmem94 in mouse model generated by CRISPR/Cas9 was embryonic lethal and led to craniofacial and cardiac abnormalities and abnormal neuronal migration pattern, suggesting that this gene is important in craniofacial, cardiovascular, and nervous system development. Our study suggests the genetic etiology of a recognizable dysmorphic syndrome with NDD and CHD and highlights the role of TMEM94 in early development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - May Christine V Malicdan
- Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA; NIH Undiagnosed Diseases Program, NHGRI and the Common Fund, National Institutes of Health, Bethesda, MD 20892, USA; Office of the Clinical Director, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
34
|
Di Donato N, Timms AE, Aldinger KA, Mirzaa GM, Bennett JT, Collins S, Olds C, Mei D, Chiari S, Carvill G, Myers CT, Rivière JB, Zaki MS, Gleeson JG, Rump A, Conti V, Parrini E, Ross ME, Ledbetter DH, Guerrini R, Dobyns WB. Analysis of 17 genes detects mutations in 81% of 811 patients with lissencephaly. Genet Med 2018; 20:1354-1364. [PMID: 29671837 PMCID: PMC6195491 DOI: 10.1038/gim.2018.8] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 12/04/2017] [Indexed: 12/20/2022] Open
Abstract
PURPOSE To estimate diagnostic yield and genotype-phenotype correlations in a cohort of 811 patients with lissencephaly or subcortical band heterotopia. METHODS We collected DNA from 756 children with lissencephaly over 30 years. Many were tested for deletion 17p13.3 and mutations of LIS1, DCX, and ARX, but few other genes. Among those tested, 216 remained unsolved and were tested by a targeted panel of 17 genes (ACTB, ACTG1, ARX, CRADD, DCX, LIS1, TUBA1A, TUBA8, TUBB2B, TUBB, TUBB3, TUBG1, KIF2A, KIF5C, DYNC1H1, RELN, and VLDLR) or by whole-exome sequencing. Fifty-five patients studied at another institution were added as a validation cohort. RESULTS The overall mutation frequency in the entire cohort was 81%. LIS1 accounted for 40% of patients, followed by DCX (23%), TUBA1A (5%), and DYNC1H1 (3%). Other genes accounted for 1% or less of patients. Nineteen percent remained unsolved, which suggests that several additional genes remain to be discovered. The majority of unsolved patients had posterior pachygyria, subcortical band heterotopia, or mild frontal pachygyria. CONCLUSION The brain-imaging pattern correlates with mutations in single lissencephaly-associated genes, as well as in biological pathways. We propose the first LIS classification system based on the underlying molecular mechanisms.
Collapse
Affiliation(s)
| | - Andrew E Timms
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Kimberly A Aldinger
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Ghayda M Mirzaa
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - James T Bennett
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Sarah Collins
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Carissa Olds
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Davide Mei
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Excellence Centre, A. Meyer Children's Hospital, University of Florence, Florence, Italy
| | - Sara Chiari
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Excellence Centre, A. Meyer Children's Hospital, University of Florence, Florence, Italy
| | - Gemma Carvill
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Candace T Myers
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Jean-Baptiste Rivière
- Department of Human Genetics, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Maha S Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Division, National Research Centre, Cairo, Egypt
| | - Joseph G Gleeson
- Department of Neurosciences, University of California San Diego, La Jolla, California, USA
| | - Andreas Rump
- Institute for Clinical Genetics, TU Dresden, Dresden, Germany
| | - Valerio Conti
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Excellence Centre, A. Meyer Children's Hospital, University of Florence, Florence, Italy
| | - Elena Parrini
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Excellence Centre, A. Meyer Children's Hospital, University of Florence, Florence, Italy
| | - M Elizabeth Ross
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, New York, USA
| | | | - Renzo Guerrini
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Excellence Centre, A. Meyer Children's Hospital, University of Florence, Florence, Italy
| | - William B Dobyns
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA.
- Department of Pediatrics, University of Washington, Seattle, Washington, USA.
- Department of Neurology, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
35
|
Calhoun JD, Carvill GL. Unravelling the genetic architecture of autosomal recessive epilepsy in the genomic era. J Neurogenet 2018; 32:295-312. [PMID: 30247086 DOI: 10.1080/01677063.2018.1513509] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The technological advancement of next-generation sequencing has greatly accelerated the pace of variant discovery in epilepsy. Despite an initial focus on autosomal dominant epilepsy due to the tractable nature of variant discovery with trios under a de novo model, more and more variants are being reported in families with epilepsies consistent with autosomal recessive (AR) inheritance. In this review, we touch on the classical AR epilepsy variants such as the inborn errors of metabolism and malformations of cortical development. However, we also highlight recently reported genes that are being identified by next-generation sequencing approaches and online 'matchmaking' platforms. Syndromes mainly characterized by seizures and complex neurodevelopmental disorders comorbid with epilepsy are discussed as an example of the wide phenotypic spectrum associated with the AR epilepsies. We conclude with a foray into the future, from the application of whole-genome sequencing to identify elusive epilepsy variants, to the promise of precision medicine initiatives to provide novel targeted therapeutics specific to the individual based on their clinical genetic testing.
Collapse
Affiliation(s)
- Jeffrey D Calhoun
- a Department of Neurology , Northwestern University Feinberg School of Medicine , Chicago , IL , USA
| | - Gemma L Carvill
- a Department of Neurology , Northwestern University Feinberg School of Medicine , Chicago , IL , USA
| |
Collapse
|
36
|
Kawasaki H. Molecular Investigations of the Development and Diseases of Cerebral Cortex Folding using Gyrencephalic Mammal Ferrets. Biol Pharm Bull 2018; 41:1324-1329. [DOI: 10.1248/bpb.b18-00142] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University
| |
Collapse
|
37
|
Tan AP, Chong WK, Mankad K. Comprehensive genotype-phenotype correlation in lissencephaly. Quant Imaging Med Surg 2018; 8:673-693. [PMID: 30211035 DOI: 10.21037/qims.2018.08.08] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Malformations of cortical development (MCD) are a heterogenous group of disorders with diverse genotypic and phenotypic variations. Lissencephaly is a subtype of MCD caused by defect in neuronal migration, which occurs between 12 and 24 weeks of gestation. The continuous advancement in the field of molecular genetics in the last decade has led to identification of at least 19 lissencephaly-related genes, most of which are related to microtubule structural proteins (tubulin) or microtubule-associated proteins (MAPs). The aim of this review article is to bring together current knowledge of gene mutations associated with lissencephaly and to provide a comprehensive genotype-phenotype correlation. Illustrative cases will be presented to facilitate the understanding of the described genotype-phenotype correlation.
Collapse
Affiliation(s)
- Ai Peng Tan
- Department of Diagnostic Imaging, National University Health System, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore
| | - Wui Khean Chong
- Department of Neuroradiology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Kshitij Mankad
- Department of Neuroradiology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
38
|
Genetics and mechanisms leading to human cortical malformations. Semin Cell Dev Biol 2018; 76:33-75. [DOI: 10.1016/j.semcdb.2017.09.031] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 09/21/2017] [Accepted: 09/21/2017] [Indexed: 02/06/2023]
|
39
|
Paspaliaris V, Vrachnis N, Iliodromiti Z, Antonakopoulos N, Papaioannou G, Vlachadis N, Anastasiadou F, Sotiriou S, Garas A, Thomaidis L, Manolakos E. 7q Deletion/12q Duplication Is the Possible Cause of an Alobar Holoprosencephaly Case. Mol Syndromol 2018; 9:52-57. [PMID: 29456484 DOI: 10.1159/000481972] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2017] [Indexed: 11/19/2022] Open
Abstract
Holoprosencephaly (HPE) spectrum disorder is the most common congenital malformation of the human brain with absence of or incomplete midline cleavage. Its cause is heterogenic, making genetic counseling a challenge. In this case report, a pregnancy affected by alobar HPE is described. Using aCGH, an 8.9-Mb deletion at 7q36.1q36.3 together with a 4.9-Mb duplication at 12q24.32q24.33 is assumed to be the possible reason for this alobar HPE case. It is discussed that disruption of key elements of the developing brain, taking environmental factors into account, contributes to the HPE spectrum. The use of aCGH for invasive prenatal testing is starting to become the standard technique, providing accurate information about the cause of congenital diseases for couples receiving genetic counseling.
Collapse
Affiliation(s)
- Vassilis Paspaliaris
- Access to genome (ATG P.C.), Clinical Laboratory Genetics, University of Athens, Athens, Greece
| | | | | | | | - Giorgos Papaioannou
- Department of Gynecology, Attikon Hospital, University of Athens, Athens, Greece
| | | | - Foteini Anastasiadou
- Department of Clinical Embryology, Larissa Medical School, University of Thessaly, Larissa, Greece
| | - Sotirios Sotiriou
- Department of Clinical Embryology, Larissa Medical School, University of Thessaly, Larissa, Greece
| | - Antonios Garas
- Department of Gynecology, Larissa Medical School, University of Thessaly, Larissa, Greece
| | - Lorreta Thomaidis
- Developmental Assessment Unit, 2nd Department of Pediatrics, P&A Kyriakou Children's Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Emmanouil Manolakos
- Access to genome (ATG P.C.), Clinical Laboratory Genetics, University of Athens, Athens, Greece.,Developmental Assessment Unit, 2nd Department of Pediatrics, P&A Kyriakou Children's Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Department of Medical Genetics, University of Cagliari, Binaghi Hospital, Cagliari, Italy
| |
Collapse
|
40
|
An Essential Postdevelopmental Role for Lis1 in Mice. eNeuro 2018; 5:eN-NWR-0350-17. [PMID: 29404402 PMCID: PMC5797476 DOI: 10.1523/eneuro.0350-17.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/13/2018] [Accepted: 01/17/2018] [Indexed: 12/15/2022] Open
Abstract
LIS1 mutations cause lissencephaly (LIS), a severe developmental brain malformation. Much less is known about its role in the mature nervous system. LIS1 regulates the microtubule motor cytoplasmic dynein 1 (dynein), and as LIS1 and dynein are both expressed in the adult nervous system, Lis1 could potentially regulate dynein-dependent processes such as axonal transport. We therefore knocked out Lis1 in adult mice using tamoxifen-induced, Cre-ER-mediated recombination. When an actin promoter was used to drive Cre-ER expression (Act-Cre-ER), heterozygous Lis1 knockout (KO) caused no obvious change in viability or behavior, despite evidence of widespread recombination by a Cre reporter three weeks after tamoxifen exposure. In contrast, homozygous Lis1 KO caused the rapid onset of neurological symptoms in both male and female mice. One tamoxifen-dosing regimen caused prominent recombination in the midbrain/hindbrain, PNS, and cardiac/skeletal muscle within a week; these mice developed severe symptoms in that time frame and were killed. A different tamoxifen regimen resulted in delayed recombination in midbrain/hindbrain, but not in other tissues, and also delayed the onset of symptoms. This indicates that Lis1 loss in the midbrain/hindbrain causes the severe phenotype. In support of this, brainstem regions known to house cardiorespiratory centers showed signs of axonal dysfunction in KO animals. Transport defects, neurofilament (NF) alterations, and varicosities were observed in axons in cultured DRG neurons from KO animals. Because no symptoms were observed when a cardiac specific Cre-ER promoter was used, we propose a vital role for Lis1 in autonomic neurons and implicate defective axonal transport in the KO phenotype.
Collapse
|
41
|
Goffinet AM. The evolution of cortical development: the synapsid-diapsid divergence. Development 2017; 144:4061-4077. [PMID: 29138289 DOI: 10.1242/dev.153908] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The cerebral cortex covers the rostral part of the brain and, in higher mammals and particularly humans, plays a key role in cognition and consciousness. It is populated with neuronal cell bodies distributed in radially organized layers. Understanding the common and lineage-specific molecular mechanisms that orchestrate cortical development and evolution are key issues in neurobiology. During evolution, the cortex appeared in stem amniotes and evolved divergently in two main branches of the phylogenetic tree: the synapsids (which led to present day mammals) and the diapsids (reptiles and birds). Comparative studies in organisms that belong to those two branches have identified some common principles of cortical development and organization that are possibly inherited from stem amniotes and regulated by similar molecular mechanisms. These comparisons have also highlighted certain essential features of mammalian cortices that are absent or different in diapsids and that probably evolved after the synapsid-diapsid divergence. Chief among these is the size and multi-laminar organization of the mammalian cortex, and the propensity to increase its area by folding. Here, I review recent data on cortical neurogenesis, neuronal migration and cortical layer formation and folding in this evolutionary perspective, and highlight important unanswered questions for future investigation.
Collapse
Affiliation(s)
- Andre M Goffinet
- University of Louvain, Avenue Mounier, 73 Box B1.73.16, B1200 Brussels, Belgium
| |
Collapse
|
42
|
Shinmyo Y, Terashita Y, Dinh Duong TA, Horiike T, Kawasumi M, Hosomichi K, Tajima A, Kawasaki H. Folding of the Cerebral Cortex Requires Cdk5 in Upper-Layer Neurons in Gyrencephalic Mammals. Cell Rep 2017; 20:2131-2143. [DOI: 10.1016/j.celrep.2017.08.024] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 06/08/2017] [Accepted: 08/03/2017] [Indexed: 02/06/2023] Open
|
43
|
Di Donato N, Chiari S, Mirzaa GM, Aldinger K, Parrini E, Olds C, Barkovich AJ, Guerrini R, Dobyns WB. Lissencephaly: Expanded imaging and clinical classification. Am J Med Genet A 2017; 173:1473-1488. [PMID: 28440899 DOI: 10.1002/ajmg.a.38245] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 03/13/2017] [Indexed: 12/17/2022]
Abstract
Lissencephaly ("smooth brain," LIS) is a malformation of cortical development associated with deficient neuronal migration and abnormal formation of cerebral convolutions or gyri. The LIS spectrum includes agyria, pachygyria, and subcortical band heterotopia. Our first classification of LIS and subcortical band heterotopia (SBH) was developed to distinguish between the first two genetic causes of LIS-LIS1 (PAFAH1B1) and DCX. However, progress in molecular genetics has led to identification of 19 LIS-associated genes, leaving the existing classification system insufficient to distinguish the increasingly diverse patterns of LIS. To address this challenge, we reviewed clinical, imaging and molecular data on 188 patients with LIS-SBH ascertained during the last 5 years, and reviewed selected archival data on another ∼1,400 patients. Using these data plus published reports, we constructed a new imaging based classification system with 21 recognizable patterns that reliably predict the most likely causative genes. These patterns do not correlate consistently with the clinical outcome, leading us to also develop a new scale useful for predicting clinical severity and outcome. Taken together, our work provides new tools that should prove useful for clinical management and genetic counselling of patients with LIS-SBH (imaging and severity based classifications), and guidance for prioritizing and interpreting genetic testing results (imaging based- classification).
Collapse
Affiliation(s)
- Nataliya Di Donato
- Institute for Clinical Genetics, Tu Dresden, Dresden, Germany.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington
| | - Sara Chiari
- Paediatric Neurology and Neurogenetics Unit and Laboratories, A. Meyer Children's Hospital, Florence, Italy
| | - Ghayda M Mirzaa
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington.,Department of Pediatrics (Genetics), University of Washington, Seattle, Washington
| | - Kimberly Aldinger
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington
| | - Elena Parrini
- Paediatric Neurology and Neurogenetics Unit and Laboratories, A. Meyer Children's Hospital, Florence, Italy
| | - Carissa Olds
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington
| | - A James Barkovich
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Renzo Guerrini
- Paediatric Neurology and Neurogenetics Unit and Laboratories, A. Meyer Children's Hospital, Florence, Italy.,IRCCS Stella Maris Foundation, Pisa, Italy
| | - William B Dobyns
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington.,Department of Pediatrics (Genetics), University of Washington, Seattle, Washington.,Department of Neurology, University of Washington, Seattle, Washington
| |
Collapse
|
44
|
Colin E, Daniel J, Ziegler A, Wakim J, Scrivo A, Haack TB, Khiati S, Denommé AS, Amati-Bonneau P, Charif M, Procaccio V, Reynier P, Aleck KA, Botto LD, Herper CL, Kaiser CS, Nabbout R, N'Guyen S, Mora-Lorca JA, Assmann B, Christ S, Meitinger T, Strom TM, Prokisch H, Miranda-Vizuete A, Hoffmann GF, Lenaers G, Bomont P, Liebau E, Bonneau D. Biallelic Variants in UBA5 Reveal that Disruption of the UFM1 Cascade Can Result in Early-Onset Encephalopathy. Am J Hum Genet 2016; 99:695-703. [PMID: 27545681 DOI: 10.1016/j.ajhg.2016.06.030] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/28/2016] [Indexed: 01/10/2023] Open
Abstract
Via whole-exome sequencing, we identified rare autosomal-recessive variants in UBA5 in five children from four unrelated families affected with a similar pattern of severe intellectual deficiency, microcephaly, movement disorders, and/or early-onset intractable epilepsy. UBA5 encodes the E1-activating enzyme of ubiquitin-fold modifier 1 (UFM1), a recently identified ubiquitin-like protein. Biochemical studies of mutant UBA5 proteins and studies in fibroblasts from affected individuals revealed that UBA5 mutations impair the process of ufmylation, resulting in an abnormal endoplasmic reticulum structure. In Caenorhabditis elegans, knockout of uba-5 and of human orthologous genes in the UFM1 cascade alter cholinergic, but not glutamatergic, neurotransmission. In addition, uba5 silencing in zebrafish decreased motility while inducing abnormal movements suggestive of seizures. These clinical, biochemical, and experimental findings support our finding of UBA5 mutations as a pathophysiological cause for early-onset encephalopathies due to abnormal protein ufmylation.
Collapse
Affiliation(s)
- Estelle Colin
- Department of Biochemistry and Genetics, University Hospital, 49933 Angers Cedex 9, France; UMR CNRS 6214-INSERM 1083 and PREMMI, University of Angers, 49933 Angers Cedex 9, France
| | - Jens Daniel
- Department of Molecular Physiology, Westfälische Wilhelms-University Münster, 48143 Münster, Germany
| | - Alban Ziegler
- Department of Biochemistry and Genetics, University Hospital, 49933 Angers Cedex 9, France; UMR CNRS 6214-INSERM 1083 and PREMMI, University of Angers, 49933 Angers Cedex 9, France
| | - Jamal Wakim
- UMR CNRS 6214-INSERM 1083 and PREMMI, University of Angers, 49933 Angers Cedex 9, France
| | - Aurora Scrivo
- Avenir-Atip team, INSERM U1051, Institute of Neurosciences of Montpellier, University of Montpellier, 34091 Montpellier Cedex 5, France
| | - Tobias B Haack
- Institute of Human Genetics, Technische Universität München, 81675 München, Germany
| | - Salim Khiati
- UMR CNRS 6214-INSERM 1083 and PREMMI, University of Angers, 49933 Angers Cedex 9, France
| | - Anne-Sophie Denommé
- Department of Biochemistry and Genetics, University Hospital, 49933 Angers Cedex 9, France; UMR CNRS 6214-INSERM 1083 and PREMMI, University of Angers, 49933 Angers Cedex 9, France
| | - Patrizia Amati-Bonneau
- Department of Biochemistry and Genetics, University Hospital, 49933 Angers Cedex 9, France; UMR CNRS 6214-INSERM 1083 and PREMMI, University of Angers, 49933 Angers Cedex 9, France
| | - Majida Charif
- UMR CNRS 6214-INSERM 1083 and PREMMI, University of Angers, 49933 Angers Cedex 9, France
| | - Vincent Procaccio
- Department of Biochemistry and Genetics, University Hospital, 49933 Angers Cedex 9, France; UMR CNRS 6214-INSERM 1083 and PREMMI, University of Angers, 49933 Angers Cedex 9, France
| | - Pascal Reynier
- Department of Biochemistry and Genetics, University Hospital, 49933 Angers Cedex 9, France; UMR CNRS 6214-INSERM 1083 and PREMMI, University of Angers, 49933 Angers Cedex 9, France
| | - Kyrieckos A Aleck
- Department of Genetics and Metabolism, Phoenix Children's Medical Group, Phoenix, AZ 85016, USA
| | - Lorenzo D Botto
- Division of Medical Genetics, Department of Pediatrics, University of Utah, Salt Lake City, UT 84132, USA
| | - Claudia Lena Herper
- Department of Molecular Physiology, Westfälische Wilhelms-University Münster, 48143 Münster, Germany
| | - Charlotte Sophia Kaiser
- Department of Molecular Physiology, Westfälische Wilhelms-University Münster, 48143 Münster, Germany
| | - Rima Nabbout
- Department of Pediatric Neurology, National Reference Center for Rare Epilepsies, University Hospital Necker-Enfants-Malades, 75015 Paris, France
| | - Sylvie N'Guyen
- Department of Pediatric Neurology, University Hospital, 49933 Angers Cedex 9, France
| | - José Antonio Mora-Lorca
- Institute of Biomedicine of Seville, University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain
| | - Birgit Assmann
- Department of General Pediatrics, Division of Pediatric Metabolic Medicine and Neuropediatrics, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Stine Christ
- Department of General Pediatrics, Division of Pediatric Metabolic Medicine and Neuropediatrics, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Technische Universität München, 81675 München, Germany; Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Tim M Strom
- Institute of Human Genetics, Technische Universität München, 81675 München, Germany; Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Holger Prokisch
- Institute of Human Genetics, Technische Universität München, 81675 München, Germany; Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Antonio Miranda-Vizuete
- Institute of Biomedicine of Seville, University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain
| | - Georg F Hoffmann
- Department of General Pediatrics, Division of Pediatric Metabolic Medicine and Neuropediatrics, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Guy Lenaers
- UMR CNRS 6214-INSERM 1083 and PREMMI, University of Angers, 49933 Angers Cedex 9, France
| | - Pascale Bomont
- Avenir-Atip team, INSERM U1051, Institute of Neurosciences of Montpellier, University of Montpellier, 34091 Montpellier Cedex 5, France
| | - Eva Liebau
- Department of Molecular Physiology, Westfälische Wilhelms-University Münster, 48143 Münster, Germany
| | - Dominique Bonneau
- Department of Biochemistry and Genetics, University Hospital, 49933 Angers Cedex 9, France; UMR CNRS 6214-INSERM 1083 and PREMMI, University of Angers, 49933 Angers Cedex 9, France.
| |
Collapse
|
45
|
Abstract
Malformations of cortical development (MCD) represent a major cause of developmental disabilities, severe epilepsy, and reproductive disadvantage. Genes that have been associated to MCD are mainly involved in cell proliferation and specification, neuronal migration, and late cortical organization. Lissencephaly-pachygyria-severe band heterotopia are diffuse neuronal migration disorders causing severe global neurological impairment. Abnormalities of the LIS1, DCX, ARX, RELN, VLDLR, ACTB, ACTG1, TUBG1, KIF5C, KIF2A, and CDK5 genes have been associated with these malformations. More recent studies have also established a relationship between lissencephaly, with or without associated microcephaly, corpus callosum dysgenesis as well as cerebellar hypoplasia, and at times, a morphological pattern consistent with polymicrogyria with mutations of several genes (TUBA1A, TUBA8, TUBB, TUBB2B, TUBB3, and DYNC1H1), regulating the synthesis and function of microtubule and centrosome key components and hence defined as tubulinopathies. MCD only affecting subsets of neurons, such as mild subcortical band heterotopia and periventricular heterotopia, have been associated with abnormalities of the DCX, FLN1A, and ARFGEF2 genes and cause neurological and cognitive impairment that vary from severe to mild deficits. Polymicrogyria results from abnormal late cortical organization and is inconstantly associated with abnormal neuronal migration. Localized polymicrogyria has been associated with anatomo-specific deficits, including disorders of language and higher cognition. Polymicrogyria is genetically heterogeneous, and only in a small minority of patients, a definite genetic cause has been identified. Megalencephaly with normal cortex or polymicrogyria by MRI imaging, hemimegalencephaly and focal cortical dysplasia can all result from mutations in genes of the PI3K-AKT-mTOR pathway. Postzygotic mutations have been described for most MCD and can be limited to the dysplastic tissue in the less diffuse forms.
Collapse
Affiliation(s)
- Elena Parrini
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Department of Neuroscience, A. Meyer Children's Hospital, University of Florence, Florence, Italy
| | - Valerio Conti
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Department of Neuroscience, A. Meyer Children's Hospital, University of Florence, Florence, Italy
| | - William B Dobyns
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, and Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Wash., USA
| | - Renzo Guerrini
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Department of Neuroscience, A. Meyer Children's Hospital, University of Florence, Florence, Italy
| |
Collapse
|
46
|
Jiang X, Nardelli J. Cellular and molecular introduction to brain development. Neurobiol Dis 2016; 92:3-17. [PMID: 26184894 PMCID: PMC4720585 DOI: 10.1016/j.nbd.2015.07.007] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 07/07/2015] [Accepted: 07/09/2015] [Indexed: 12/13/2022] Open
Abstract
Advances in the study of brain development over the last decades, especially recent findings regarding the evolutionary expansion of the human neocortex, and large-scale analyses of the proteome/transcriptome in the human brain, have offered novel insights into the molecular mechanisms guiding neural maturation, and the pathophysiology of multiple forms of neurological disorders. As a preamble to reviews of this issue, we provide an overview of the cellular, molecular and genetic bases of brain development with an emphasis on the major mechanisms associated with landmarks of normal neural development in the embryonic stage and early postnatal life, including neural stem/progenitor cell proliferation, cortical neuronal migration, evolution and folding of the cerebral cortex, synaptogenesis and neural circuit development, gliogenesis and myelination. We will only briefly depict developmental disorders that result from perturbations of these cellular or molecular mechanisms, and the most common perinatal brain injuries that could disturb normal brain development.
Collapse
Affiliation(s)
- Xiangning Jiang
- Department of Pediatrics, University of California, San Francisco, CA 94158, USA
| | - Jeannette Nardelli
- Inserm, U1141, Paris 75019, France; Université Paris Diderot, Sorbonne Paris Cité, UMRS 1141, Paris 75019, France.
| |
Collapse
|
47
|
Functional characterization of CDK5 and CDK5R1 mutations identified in patients with non-syndromic intellectual disability. J Hum Genet 2015; 61:283-93. [PMID: 26657932 DOI: 10.1038/jhg.2015.144] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 10/27/2015] [Accepted: 11/06/2015] [Indexed: 12/13/2022]
Abstract
Cyclin-dependent kinase 5 (CDK5) and cyclin-dependent kinase 5, regulatory subunit 1 (CDK5R1), encoding CDK5 activator p35, have a fundamental role in central nervous system (CNS) development and function, and are involved in the pathogenesis of several neurodegenerative disorders, thus constituting strong candidate genes for the onset of intellectual disability (ID). We carried out a mutation screening of CDK5 and CDK5R1 coding regions and CDK5R1 3'-UTR on a cohort of 360 patients with non-syndromic ID (NS-ID) using denaturing high performance liquid chromatography (DHPLC) and direct sequencing. We found one novel silent mutation in CDK5 and one novel silent mutation in CDK5R1 coding regions, three novel intronic variations in CDK5, not causing any splicing defect, and four novel heterozygous variations in CDK5R1 3'-UTR. None of these variations was present in 450 healthy controls and single-nucleotide polymorphism (SNP) databases. The functional study of CDK5R1 p.A108V mutation evidenced an impaired p35 cleavage by the calcium-dependent protease calpain. Moreover, luciferase constructs containing the CDK5R1 3'-UTR mutations showed altered gene expression levels. Eight known polymorphisms were also identified displaying different frequencies in NS-ID patients compared with the controls. In particular, the minor allele of CDK5R1 3'-UTR rs735555 polymorphism was associated with increased risk for NS-ID. In conclusion, our data suggest that mutations and polymorphisms in CDK5 and CDK5R1 genes may contribute to the onset of the NS-ID phenotype.
Collapse
|
48
|
Takada S, Mizuno K, Saito T, Asada A, Giese KP, Hisanaga SI. Effects of p35 Mutations Associated with Mental Retardation on the Cellular Function of p35-CDK5. PLoS One 2015; 10:e0140821. [PMID: 26469698 PMCID: PMC4607440 DOI: 10.1371/journal.pone.0140821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/29/2015] [Indexed: 12/02/2022] Open
Abstract
p35 is an activation subunit of the cyclin-dependent kinase 5 (CDK5), which is a Ser/Thr kinase that is expressed predominantly in neurons. Disruption of the CDK5 or p35 (CDK5R1) genes induces abnormal neuronal layering in various regions of the mouse brain via impaired neuronal migration, which may be relevant for mental retardation in humans. Accordingly, mutations in the p35 gene were reported in patients with nonsyndromic mental retardation; however, their effect on the biochemical function of p35 has not been examined. Here, we studied the biochemical effect of mutant p35 on its known properties, i.e., stability, CDK5 activation, and cellular localization, using heterologous expression in cultured cells. We also examined the effect of the mutations on axon elongation in cultured primary neurons and migration of newborn neurons in embryonic brains. However, we did not detect any significant differences in the effects of the mutant forms of p35 compared with wild-type p35. Therefore, we conclude that these p35 mutations are unlikely to cause mental retardation.
Collapse
Affiliation(s)
- Shunsuke Takada
- Laboratory of Molecular Neuroscience, Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Minami-Osawa, Hachioji, Tokyo, Japan
| | - Keiko Mizuno
- Centre for Cellular Basis of Behavior, Institute of Psychiatry, King's College London, 125 Coldharbour Lane, London, United Kingdom
| | - Taro Saito
- Laboratory of Molecular Neuroscience, Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Minami-Osawa, Hachioji, Tokyo, Japan
| | - Akiko Asada
- Laboratory of Molecular Neuroscience, Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Minami-Osawa, Hachioji, Tokyo, Japan
| | - Karl Peter Giese
- Centre for Cellular Basis of Behavior, Institute of Psychiatry, King's College London, 125 Coldharbour Lane, London, United Kingdom
| | - Shin-ichi Hisanaga
- Laboratory of Molecular Neuroscience, Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Minami-Osawa, Hachioji, Tokyo, Japan
- * E-mail:
| |
Collapse
|
49
|
Kawauchi T. Cellullar insights into cerebral cortical development: focusing on the locomotion mode of neuronal migration. Front Cell Neurosci 2015; 9:394. [PMID: 26500496 PMCID: PMC4595654 DOI: 10.3389/fncel.2015.00394] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 09/22/2015] [Indexed: 02/01/2023] Open
Abstract
The mammalian brain consists of numerous compartments that are closely connected with each other via neural networks, comprising the basis of higher order brain functions. The highly specialized structure originates from simple pseudostratified neuroepithelium-derived neural progenitors located near the ventricle. A long journey by neurons from the ventricular side is essential for the formation of a sophisticated brain structure, including a mammalian-specific six-layered cerebral cortex. Neuronal migration consists of several contiguous steps, but the locomotion mode comprises a large part of the migration. The locomoting neurons exhibit unique features; a radial glial fiber-dependent migration requiring the endocytic recycling of N-cadherin and a neuron-specific migration mode with dilation/swelling formation that requires the actin and microtubule organization possibly regulated by cyclin-dependent kinase 5 (Cdk5), Dcx, p27(kip1), Rac1, and POSH. Here I will introduce the roles of various cellular events, such as cytoskeletal organization, cell adhesion, and membrane trafficking, in the regulation of the neuronal migration, with particular focus on the locomotion mode.
Collapse
Affiliation(s)
- Takeshi Kawauchi
- Department of Physiology, Keio University School of Medicine Tokyo, Japan ; Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency Saitama, Japan ; Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation Kobe, Japan
| |
Collapse
|