51
|
Passemard S, Verloes A, Billette de Villemeur T, Boespflug-Tanguy O, Hernandez K, Laurent M, Isidor B, Alberti C, Pouvreau N, Drunat S, Gérard B, El Ghouzzi V, Gallego J, Elmaleh-Bergès M, Huttner WB, Eliez S, Gressens P, Schaer M. Abnormal spindle-like microcephaly-associated (ASPM) mutations strongly disrupt neocortical structure but spare the hippocampus and long-term memory. Cortex 2016; 74:158-76. [DOI: 10.1016/j.cortex.2015.10.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 09/14/2015] [Accepted: 10/19/2015] [Indexed: 01/21/2023]
|
52
|
Species-Specific Expression of Full-Length and Alternatively Spliced Variant Forms of CDK5RAP2. PLoS One 2015; 10:e0142577. [PMID: 26550838 PMCID: PMC4638350 DOI: 10.1371/journal.pone.0142577] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 10/24/2015] [Indexed: 11/30/2022] Open
Abstract
CDK5RAP2 is one of the primary microcephaly genes that are associated with reduced brain size and mental retardation. We have previously shown that human CDK5RAP2 exists as a full-length form (hCDK5RAP2) or an alternatively spliced variant form (hCDK5RAP2-V1) that is lacking exon 32. The equivalent of hCDK5RAP2-V1 has been reported in rat and mouse but the presence of full-length equivalent hCDK5RAP2 in rat and mouse has not been examined. Here, we demonstrate that rat expresses both a full length and an alternatively spliced variant form of CDK5RAP2 that are equivalent to our previously reported hCDK5RAP2 and hCDK5RAP2-V1, repectively. However, mouse expresses only one form of CDK5RAP2 that is equivalent to the human and rat alternatively spliced variant forms. Knowledge of this expression of different forms of CDK5RAP2 in human, rat and mouse is essential in selecting the appropriate model for studies of CDK5RAP2 and primary microcephaly but our findings further indicate the evolutionary divergence of mouse from the human and rat species.
Collapse
|
53
|
Pan X, Chang X, Leung C, Zhou Z, Cao F, Xie W, Jia Z. PAK1 regulates cortical development via promoting neuronal migration and progenitor cell proliferation. Mol Brain 2015; 8:36. [PMID: 26043730 PMCID: PMC4456803 DOI: 10.1186/s13041-015-0124-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/12/2015] [Indexed: 11/17/2022] Open
Abstract
Background p21-activated kinase 1 (PAK1) is a serine/threonine kinase known to be activated by the Rho family small GTPases and to play a key role in cytoskeletal reorganization, spine morphology and synaptic plasticity. PAK1 is also implicated in a number of neurodevelopmental and neurodegenerative diseases, including autism, intellectual disability and Alzheimer’s disease. However, the role of PAK1 in early brain development remains unknown. Results In this study, we employed genetic manipulations to investigate the role of PAK1 in the cerebral cortical development in mice. We showed that compared to the wild type littermates, PAK1 knockout mice have a reduction in the number of pyramidal neurons in several layers of the cerebral cortex, which is associated with a smaller pool of neural progenitor cells and impaired neuronal migration. Conclusion These results suggest that PAK1 regulates cortical development by promoting the proliferation of neural progenitor cells and facilitating the migration of these neurons to specific regions of the cortex.
Collapse
Affiliation(s)
- Xingxiu Pan
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, 2 Sipailou Road, 210096, Nanjing, China.
| | - Xinxia Chang
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, 2 Sipailou Road, 210096, Nanjing, China.
| | - Celeste Leung
- Neurosciences & Mental Health, The Hospital for Sick Children, 555 University Ave., M5G 1X8, Toronto, Ontario, Canada. .,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada.
| | - Zikai Zhou
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, 2 Sipailou Road, 210096, Nanjing, China.
| | - Feng Cao
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, 2 Sipailou Road, 210096, Nanjing, China. .,Neurosciences & Mental Health, The Hospital for Sick Children, 555 University Ave., M5G 1X8, Toronto, Ontario, Canada. .,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada.
| | - Wei Xie
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, 2 Sipailou Road, 210096, Nanjing, China.
| | - Zhengping Jia
- Neurosciences & Mental Health, The Hospital for Sick Children, 555 University Ave., M5G 1X8, Toronto, Ontario, Canada. .,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada.
| |
Collapse
|
54
|
Morris-Rosendahl DJ, Kaindl AM. What next-generation sequencing (NGS) technology has enabled us to learn about primary autosomal recessive microcephaly (MCPH). Mol Cell Probes 2015; 29:271-81. [PMID: 26050940 DOI: 10.1016/j.mcp.2015.05.015] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 05/28/2015] [Accepted: 05/29/2015] [Indexed: 11/27/2022]
Abstract
The impact that next-generation sequencing technology (NGS) is having on many aspects of molecular and cell biology, is becoming increasingly apparent. One of the most noticeable outcomes of the new technology in human genetics, has been the accelerated rate of identification of disease-causing genes. Especially for rare, heterogeneous disorders, such as autosomal recessive primary microcephaly (MCPH), the handful of genes previously known to harbour disease-causing mutations, has grown at an unprecedented rate within a few years. Knowledge of new genes mutated in MCPH over the last four years has contributed to our understanding of the disorder at both the clinical and cellular levels. The functions of proteins such as WDR62, CASC5, PHC1, CDK6, CENP-E, CENP-F, CEP63, ZNF335, PLK4 and TUBGPC, have been added to the complex network of critical cellular processes known to be involved in brain growth and size. In addition to the importance of mitotic spindle assembly and structure, centrosome and centriole function and DNA repair and damage response, new mechanisms involving kinetochore-associated proteins and chromatin remodelling complexes have been elucidated. Two of the major contributions to our clinical knowledge are the realisation that primary microcephaly caused by mutations in genes at the MCPH loci is seldom an isolated clinical feature and is often accompanied either by additional cortical malformations or primordial dwarfism. Gene-phenotype correlations are being revisited, with a new dimension of locus heterogeneity and phenotypic variability being revealed.
Collapse
Affiliation(s)
- Deborah J Morris-Rosendahl
- Clinical Genetics and Genomics, Royal Brompton and Harefield NHS Foundation Trust, London, United Kingdom; National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| | - Angela M Kaindl
- Department of Pediatric Neurology, Charité University Medicine, Berlin, Germany; Institute of Cell Biology and Neurobiology, Charité University Medicine, Berlin, Germany
| |
Collapse
|
55
|
Saha S, Woodbine L, Haines J, Coster M, Ricket N, Barazzuol L, Ainsbury E, Sienkiewicz Z, Jeggo P. Increased apoptosis and DNA double-strand breaks in the embryonic mouse brain in response to very low-dose X-rays but not 50 Hz magnetic fields. J R Soc Interface 2015; 11:20140783. [PMID: 25209403 PMCID: PMC4191111 DOI: 10.1098/rsif.2014.0783] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The use of X-rays for medical diagnosis is enhancing exposure to low radiation doses. Exposure to extremely low-frequency electromagnetic or magnetic fields is also increasing. Epidemiological studies show consistent associations of childhood leukaemia with exposure to magnetic fields but any causal relationship is unclear. A limitation in assessing the consequence of such exposure is the availability of sensitive assays. The embryonic neuronal stem and progenitor cell compartments are radiosensitive tissues. Using sensitive assays, we report a statistically significant increase in DNA double-strand break (DSB) formation and apoptosis in the embryonic neuronal stem cell compartment following in utero exposure to 10–200 mGy X-rays. Both endpoints show a linear response. We also show that DSB repair is delayed following exposure to doses below 50 mGy compared with 100 mGy. Thus, we demonstrate in vivo consequences of low-dose radiation. In contrast to these impacts, we did not observe any significant induction of DSBs or apoptosis following exposure to 50 Hz magnetic fields (100 or 300 µT). We conclude that any DSB induction by treatment with magnetic fields is lower than following exposure to 10 mGy X-rays. For comparison, certain procedures involving computed tomography scanning are equivalent to 1–5 mGy X-rays.
Collapse
Affiliation(s)
- Shreya Saha
- Genome Damage and Stability Centre, Life Sciences, University of Sussex, Brighton BN19RQ, UK
| | - Lisa Woodbine
- Genome Damage and Stability Centre, Life Sciences, University of Sussex, Brighton BN19RQ, UK
| | - Jackie Haines
- Public Health England Centre for Radiation, Chemical and Environmental Hazards, Chilton, Didcot, Oxford OX11 0RQ, UK
| | - Margaret Coster
- Public Health England Centre for Radiation, Chemical and Environmental Hazards, Chilton, Didcot, Oxford OX11 0RQ, UK
| | - Nicole Ricket
- Genome Damage and Stability Centre, Life Sciences, University of Sussex, Brighton BN19RQ, UK
| | - Lara Barazzuol
- Genome Damage and Stability Centre, Life Sciences, University of Sussex, Brighton BN19RQ, UK
| | - Elizabeth Ainsbury
- Public Health England Centre for Radiation, Chemical and Environmental Hazards, Chilton, Didcot, Oxford OX11 0RQ, UK
| | - Zenon Sienkiewicz
- Public Health England Centre for Radiation, Chemical and Environmental Hazards, Chilton, Didcot, Oxford OX11 0RQ, UK
| | - Penny Jeggo
- Genome Damage and Stability Centre, Life Sciences, University of Sussex, Brighton BN19RQ, UK
| |
Collapse
|
56
|
Chromosome structure deficiencies in MCPH1 syndrome. Chromosoma 2015; 124:491-501. [PMID: 25845520 DOI: 10.1007/s00412-015-0512-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 03/06/2015] [Accepted: 03/16/2015] [Indexed: 01/11/2023]
Abstract
Mutations in the MCPH1 gene result in primary microcephaly in combination with a unique cellular phenotype of defective chromosome condensation. MCPH1 patient cells display premature chromosome condensation in G2 phase of the cell cycle and delayed decondensation in early G1 phase, observable as an increased proportion of cells with prophase-like appearance. MCPH1 deficiency thus appears to uncouple the chromosome cycle from the coordinated series of events that take place during mitosis such as some phases of the centrosome cycle and nuclear envelope breakdown. Here, we provide a further characterization of the effects of MCPH1 loss-of-function on chromosome morphology. In comparison to healthy controls, chromosomes of MCPH1 patients are shorter and display a pronounced coiling of their central chromatid axes. In addition, a substantial fraction of metaphase chromosomes shows apparently unresolved chromatids with twisted appearance. The patient chromosomes also showed signs of defective centromeric cohesion, which become more apparent and pronounced after harsh hypotonic conditions. Taking together, the observed alterations indicate additional so far unknown functions of MCPH1 during chromosome shaping and dynamics.
Collapse
|
57
|
Ito H, Shiwaku H, Yoshida C, Homma H, Luo H, Chen X, Fujita K, Musante L, Fischer U, Frints SGM, Romano C, Ikeuchi Y, Shimamura T, Imoto S, Miyano S, Muramatsu SI, Kawauchi T, Hoshino M, Sudol M, Arumughan A, Wanker EE, Rich T, Schwartz C, Matsuzaki F, Bonni A, Kalscheuer VM, Okazawa H. In utero gene therapy rescues microcephaly caused by Pqbp1-hypofunction in neural stem progenitor cells. Mol Psychiatry 2015; 20:459-71. [PMID: 25070536 PMCID: PMC4378255 DOI: 10.1038/mp.2014.69] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 04/10/2014] [Accepted: 05/12/2014] [Indexed: 12/21/2022]
Abstract
Human mutations in PQBP1, a molecule involved in transcription and splicing, result in a reduced but architecturally normal brain. Examination of a conditional Pqbp1-knockout (cKO) mouse with microcephaly failed to reveal either abnormal centrosomes or mitotic spindles, increased neurogenesis from the neural stem progenitor cell (NSPC) pool or increased cell death in vivo. Instead, we observed an increase in the length of the cell cycle, particularly for the M phase in NSPCs. Corresponding to the developmental expression of Pqbp1, the stem cell pool in vivo was decreased at E10 and remained at a low level during neurogenesis (E15) in Pqbp1-cKO mice. The expression profiles of NSPCs derived from the cKO mouse revealed significant changes in gene groups that control the M phase, including anaphase-promoting complex genes, via aberrant transcription and RNA splicing. Exogenous Apc4, a hub protein in the network of affected genes, recovered the cell cycle, proliferation, and cell phenotypes of NSPCs caused by Pqbp1-cKO. These data reveal a mechanism of brain size control based on the simple reduction of the NSPC pool by cell cycle time elongation. Finally, we demonstrated that in utero gene therapy for Pqbp1-cKO mice by intraperitoneal injection of the PQBP1-AAV vector at E10 successfully rescued microcephaly with preserved cortical structures and improved behavioral abnormalities in Pqbp1-cKO mice, opening a new strategy for treating this intractable developmental disorder.
Collapse
Affiliation(s)
- H Ito
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - H Shiwaku
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - C Yoshida
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - H Homma
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - H Luo
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - X Chen
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - K Fujita
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - L Musante
- Department of Human Molecular Genetics, Max-Planck Institute for Molecular Genetics, Berlin-Dahlem, Germany
| | - U Fischer
- Department of Human Molecular Genetics, Max-Planck Institute for Molecular Genetics, Berlin-Dahlem, Germany
| | - S G M Frints
- Department of Clinical Genetics, University Hospital azM Maastricht, Maastricht, The Netherlands,School for Oncology and Developmental Biology, GROW, Maastricht University, Maastricht, The Netherlands
| | - C Romano
- Unità Operativa Complessa di Pediatria e Genetica Medica, IRCCS Associazione Oasi Maria Santissima, Troina (Enna), Italy
| | - Y Ikeuchi
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St Louis, MO, USA,Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - T Shimamura
- Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - S Imoto
- Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - S Miyano
- Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - S-i Muramatsu
- Department of Neurology, Jichi Medical University, Tochigi, Japan
| | - T Kawauchi
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - M Hoshino
- Department of Biochemistry and Cellular Biology, National Center for Neurology and Psychiatry, Tokyo, Japan
| | - M Sudol
- Laboratory of Signal Transduction and Proteomic Profiling, Weis Center for Research, Geisinger Clinic, Danville, PA, USA
| | - A Arumughan
- Department of Neurogenetics, Max-Delbrück Center for Molecular Medicine, Berlin-Buch, Germany
| | - E E Wanker
- Department of Neurogenetics, Max-Delbrück Center for Molecular Medicine, Berlin-Buch, Germany
| | - T Rich
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - C Schwartz
- JC Self Research Institute of Human Genetics, Greenwood Genetic Center, Greenwood, SC, USA
| | - F Matsuzaki
- Laboratory for Cell Asymmetry, Center for Developmental Biology, RIKEN, Chuo-ku, Kobe, Japan
| | - A Bonni
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St Louis, MO, USA,Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - V M Kalscheuer
- Department of Human Molecular Genetics, Max-Planck Institute for Molecular Genetics, Berlin-Dahlem, Germany
| | - H Okazawa
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan,Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan. E-mail:
| |
Collapse
|
58
|
Woodbine L, Haines J, Coster M, Barazzuol L, Ainsbury E, Sienkiewicz Z, Jeggo P. The rate of X-ray-induced DNA double-strand break repair in the embryonic mouse brain is unaffected by exposure to 50 Hz magnetic fields. Int J Radiat Biol 2015; 91:495-9. [PMID: 25786477 PMCID: PMC4673581 DOI: 10.3109/09553002.2015.1021963] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Purpose: Following in utero exposure to low dose radiation (10–200 mGy), we recently observed a linear induction of DNA double-strand breaks (DSB) and activation of apoptosis in the embryonic neuronal stem/progenitor cell compartment. No significant induction of DSB or apoptosis was observed following exposure to magnetic fields (MF). In the present study, we exploited this in vivo system to examine whether exposure to MF before and after exposure to 100 mGy X-rays impacts upon DSB repair rates. Materials and methods: 53BP1 foci were quantified following combined exposure to radiation and MF in the embryonic neuronal stem/progenitor cell compartment. Embryos were exposed in utero to 50 Hz MF at 300 μT for 3 h before and up to 9 h after exposure to 100 mGy X-rays. Controls included embryos exposed to MF or X-rays alone plus sham exposures. Results: Exposure to MF before and after 100 mGy X-rays did not impact upon the rate of DSB repair in the embryonic neuronal stem cell compartment compared to repair rates following radiation exposure alone. Conclusions: We conclude that in this sensitive system MF do not exert any significant level of DNA damage and do not impede the repair of X-ray induced damage.
Collapse
Affiliation(s)
- Lisa Woodbine
- Genome Damage and Stability Centre, Life Sciences, University of Sussex , Brighton
| | | | | | | | | | | | | |
Collapse
|
59
|
Faheem M, Naseer MI, Rasool M, Chaudhary AG, Kumosani TA, Ilyas AM, Pushparaj P, Ahmed F, Algahtani HA, Al-Qahtani MH, Saleh Jamal H. Molecular genetics of human primary microcephaly: an overview. BMC Med Genomics 2015; 8 Suppl 1:S4. [PMID: 25951892 PMCID: PMC4315316 DOI: 10.1186/1755-8794-8-s1-s4] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Autosomal recessive primary microcephaly (MCPH) is a neurodevelopmental disorder that is characterised by microcephaly present at birth and non-progressive mental retardation. Microcephaly is the outcome of a smaller but architecturally normal brain; the cerebral cortex exhibits a significant decrease in size. MCPH is a neurogenic mitotic disorder, though affected patients demonstrate normal neuronal migration, neuronal apoptosis and neural function. Twelve MCPH loci (MCPH1-MCPH12) have been mapped to date from various populations around the world and contain the following genes: Microcephalin, WDR62, CDK5RAP2, CASC5, ASPM, CENPJ, STIL, CEP135, CEP152, ZNF335, PHC1 and CDK6. It is predicted that MCPH gene mutations may lead to the disease phenotype due to a disturbed mitotic spindle orientation, premature chromosomal condensation, signalling response as a result of damaged DNA, microtubule dynamics, transcriptional control or a few other hidden centrosomal mechanisms that can regulate the number of neurons produced by neuronal precursor cells. Additional findings have further elucidated the microcephaly aetiology and pathophysiology, which has informed the clinical management of families suffering from MCPH. The provision of molecular diagnosis and genetic counselling may help to decrease the frequency of this disorder.
Collapse
|
60
|
Milovanova OA. Cortical dysgenesis with epileptic syndromes and symptomatic epilepsy in children. Zh Nevrol Psikhiatr Im S S Korsakova 2015; 115:154-161. [DOI: 10.17116/jnevro2015115112154-161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
61
|
Molecular and cellular basis of autosomal recessive primary microcephaly. BIOMED RESEARCH INTERNATIONAL 2014; 2014:547986. [PMID: 25548773 PMCID: PMC4274849 DOI: 10.1155/2014/547986] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 09/18/2014] [Accepted: 09/18/2014] [Indexed: 01/23/2023]
Abstract
Autosomal recessive primary microcephaly (MCPH) is a rare hereditary neurodevelopmental disorder characterized by a marked reduction in brain size and intellectual disability. MCPH is genetically heterogeneous and can exhibit additional clinical features that overlap with related disorders including Seckel syndrome, Meier-Gorlin syndrome, and microcephalic osteodysplastic dwarfism. In this review, we discuss the key proteins mutated in MCPH. To date, MCPH-causing mutations have been identified in twelve different genes, many of which encode proteins that are involved in cell cycle regulation or are present at the centrosome, an organelle crucial for mitotic spindle assembly and cell division. We highlight recent findings on MCPH proteins with regard to their role in cell cycle progression, centrosome function, and early brain development.
Collapse
|
62
|
|
63
|
Rupp V, Rauf S, Naveed I, Windpassinger C, Mir A. A novel single base pair duplication in WDR62 causes primary microcephaly. BMC MEDICAL GENETICS 2014; 15:107. [PMID: 25303973 PMCID: PMC4258795 DOI: 10.1186/s12881-014-0107-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 09/10/2014] [Indexed: 12/19/2022]
Abstract
BACKGROUND Primary microcephaly is a disorder of the brain resulting in a reduced head circumference that can come along with intellectual disability but with hardly any other neurological abnormalities. CASE PRESENTATION In this study we report on three Pakistani males from a consanguineous family with 2, 4 and 25 years, diagnosed with autosomal recessive primary microcephaly. By genotyping, Sanger sequencing and using bioinformatical approaches the disease causing mutation was identified and evaluated. CONCLUSION By using a 250K SNP array, we were able to detect an 11Mb large autozygous region in the MCPH2 locus on chromosome 19q13.12. Sequencing of the associated gene, WDR62, revealed the frameshift causing single base pair duplication, c.2527dupG. This mutation is predicted to affect the structural features of WDR62 which in turn changes the conformation and function of the protein. Aspartic acid (D) at position 843 was found to be conserved among various ortholog species. The present findings will be helpful in genetic diagnosis of patients and future studies of WDR62.
Collapse
|
64
|
Keeney JG, Dumas L, Sikela JM. The case for DUF1220 domain dosage as a primary contributor to anthropoid brain expansion. Front Hum Neurosci 2014; 8:427. [PMID: 25009482 PMCID: PMC4067907 DOI: 10.3389/fnhum.2014.00427] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 05/28/2014] [Indexed: 12/14/2022] Open
Abstract
Here we present the hypothesis that increasing copy number (dosage) of sequences encoding DUF1220 protein domains is a major contributor to the evolutionary increase in brain size, neuron number, and cognitive capacity that is associated with the primate order. We further propose that this relationship is restricted to the anthropoid sub-order of primates, with DUF1220 copy number markedly increasing in monkeys, further in apes, and most extremely in humans where the greatest number of copies (~272 haploid copies) is found. We show that this increase closely parallels the increase in brain size and neuron number that has occurred among anthropoid primate species. We also provide evidence linking DUF1220 copy number to brain size within the human species, both in normal populations and in individuals associated with brain size pathologies (1q21-associated microcephaly and macrocephaly). While we believe these and other findings presented here strongly suggest increase in DUF1220 copy number is a key contributor to anthropoid brain expansion, the data currently available rely largely on correlative measures that, though considerable, do not yet provide direct evidence for a causal connection. Nevertheless, we believe the evidence presented is sufficient to provide the basis for a testable model which proposes that DUF1220 protein domain dosage increase is a main contributor to the increase in brain size and neuron number found among the anthropoid primate species and that is at its most extreme in human.
Collapse
Affiliation(s)
- Jonathon G Keeney
- Department of Biochemistry and Molecular Genetics and Human Medical Genetics and Neuroscience Programs, University of Colorado School of Medicine, Anschutz Medical Campus Aurora, CO, USA
| | - Laura Dumas
- Department of Biochemistry and Molecular Genetics and Human Medical Genetics and Neuroscience Programs, University of Colorado School of Medicine, Anschutz Medical Campus Aurora, CO, USA
| | - James M Sikela
- Department of Biochemistry and Molecular Genetics and Human Medical Genetics and Neuroscience Programs, University of Colorado School of Medicine, Anschutz Medical Campus Aurora, CO, USA
| |
Collapse
|
65
|
Khan MA, Rupp VM, Orpinell M, Hussain MS, Altmüller J, Steinmetz MO, Enzinger C, Thiele H, Höhne W, Nürnberg G, Baig SM, Ansar M, Nürnberg P, Vincent JB, Speicher MR, Gönczy P, Windpassinger C. A missense mutation in the PISA domain of HsSAS-6 causes autosomal recessive primary microcephaly in a large consanguineous Pakistani family. Hum Mol Genet 2014; 23:5940-9. [DOI: 10.1093/hmg/ddu318] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
66
|
Montgomery SH, Mundy NI. Microcephaly genes evolved adaptively throughout the evolution of eutherian mammals. BMC Evol Biol 2014; 14:120. [PMID: 24898820 PMCID: PMC4055943 DOI: 10.1186/1471-2148-14-120] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 05/23/2014] [Indexed: 10/27/2022] Open
Abstract
BACKGROUND Genes associated with the neurodevelopmental disorder microcephaly display a strong signature of adaptive evolution in primates. Comparative data suggest a link between selection on some of these loci and the evolution of primate brain size. Whether or not either positive selection or this phenotypic association are unique to primates is unclear, but recent studies in cetaceans suggest at least two microcephaly genes evolved adaptively in other large brained mammalian clades. RESULTS Here we analyse the evolution of seven microcephaly loci, including three recently identified loci, across 33 eutherian mammals. We find extensive evidence for positive selection having acted on the majority of these loci not just in primates but also across non-primate mammals. Furthermore, the patterns of selection in major mammalian clades are not significantly different. Using phylogenetically corrected comparative analyses, we find that the evolution of two microcephaly loci, ASPM and CDK5RAP2, are correlated with neonatal brain size in Glires and Euungulata, the two most densely sampled non-primate clades. CONCLUSIONS Together with previous results, this suggests that ASPM and CDK5RAP2 may have had a consistent role in the evolution of brain size in mammals. Nevertheless, several limitations of currently available data and gene-phenotype tests are discussed, including sparse sampling across large evolutionary distances, averaging gene-wide rates of evolution, potential phenotypic variation and evolutionary reversals. We discuss the implications of our results for studies of the genetic basis of brain evolution, and explicit tests of gene-phenotype hypotheses.
Collapse
Affiliation(s)
- Stephen H Montgomery
- Department Genetics, Evolution & Environment, University College London, Gower Street, London WC1E 6BT, UK.
| | | |
Collapse
|
67
|
Hattori N. Cerebral organoids model human brain development and microcephaly. Mov Disord 2013; 29:185. [PMID: 24375826 DOI: 10.1002/mds.25740] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 10/11/2013] [Indexed: 11/11/2022] Open
Affiliation(s)
- Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
68
|
Lancaster MA, Renner M, Martin CA, Wenzel D, Bicknell LS, Hurles ME, Homfray T, Penninger JM, Jackson AP, Knoblich JA. Cerebral organoids model human brain development and microcephaly. Nature 2013; 501:373-9. [PMID: 23995685 PMCID: PMC3817409 DOI: 10.1038/nature12517] [Citation(s) in RCA: 3456] [Impact Index Per Article: 288.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 08/02/2013] [Indexed: 02/06/2023]
Abstract
The complexity of the human brain has made it difficult to study many brain disorders in model organisms, highlighting the need for an in vitro model of human brain development. Here we have developed a human pluripotent stem cell-derived three-dimensional organoid culture system, termed cerebral organoids, that develop various discrete, although interdependent, brain regions. These include a cerebral cortex containing progenitor populations that organize and produce mature cortical neuron subtypes. Furthermore, cerebral organoids are shown to recapitulate features of human cortical development, namely characteristic progenitor zone organization with abundant outer radial glial stem cells. Finally, we use RNA interference and patient-specific induced pluripotent stem cells to model microcephaly, a disorder that has been difficult to recapitulate in mice. We demonstrate premature neuronal differentiation in patient organoids, a defect that could help to explain the disease phenotype. Together, these data show that three-dimensional organoids can recapitulate development and disease even in this most complex human tissue.
Collapse
Affiliation(s)
- Madeline A Lancaster
- Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna 1030, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Dediu D, Levinson SC. On the antiquity of language: the reinterpretation of Neandertal linguistic capacities and its consequences. Front Psychol 2013; 4:397. [PMID: 23847571 PMCID: PMC3701805 DOI: 10.3389/fpsyg.2013.00397] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 06/12/2013] [Indexed: 11/13/2022] Open
Abstract
It is usually assumed that modern language is a recent phenomenon, coinciding with the emergence of modern humans themselves. Many assume as well that this is the result of a single, sudden mutation giving rise to the full "modern package." However, we argue here that recognizably modern language is likely an ancient feature of our genus pre-dating at least the common ancestor of modern humans and Neandertals about half a million years ago. To this end, we adduce a broad range of evidence from linguistics, genetics, paleontology, and archaeology clearly suggesting that Neandertals shared with us something like modern speech and language. This reassessment of the antiquity of modern language, from the usually quoted 50,000-100,000 years to half a million years, has profound consequences for our understanding of our own evolution in general and especially for the sciences of speech and language. As such, it argues against a saltationist scenario for the evolution of language, and toward a gradual process of culture-gene co-evolution extending to the present day. Another consequence is that the present-day linguistic diversity might better reflect the properties of the design space for language and not just the vagaries of history, and could also contain traces of the languages spoken by other human forms such as the Neandertals.
Collapse
Affiliation(s)
- Dan Dediu
- Language and Genetics Department, Max Planck Institute for PsycholinguisticsNijmegen, Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University NijmegenNijmegen, Netherlands
| | - Stephen C. Levinson
- Donders Institute for Brain, Cognition and Behaviour, Radboud University NijmegenNijmegen, Netherlands
- Language and Cognition Department, Max Planck Institute for PsycholinguisticsNijmegen, Netherlands
| |
Collapse
|
70
|
Tan CA, del Gaudio D, Dempsey MA, Arndt K, Botes S, Reeder A, Das S. Analysis of ASPM in an ethnically diverse cohort of 400 patient samples: perspectives of the molecular diagnostic laboratory. Clin Genet 2013; 85:353-8. [PMID: 23611254 DOI: 10.1111/cge.12172] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 04/17/2013] [Accepted: 04/17/2013] [Indexed: 11/27/2022]
Abstract
Primary Autosomal Recessive Microcephaly (MCPH) is characterized by congenital microcephaly usually without additional clinical findings. The most common gene implicated in MCPH is ASPM and a large percentage of mutations described have been homozygous and in consanguineous families primarily of East Asian and Middle Eastern origin. ASPM sequencing was performed on 400 patients between the years 2009 and 2012. Seventy of the patient samples were also analyzed for copy number changes in the ASPM gene. Forty protein truncating mutations, including 29 novel mutations, were identified in 39 patients with MCPH. Approximately one third of patients were compound heterozygotes, indicative of non-consanguinity in these patients. In addition, 46 non-synonymous variants were identified and interpreted as variants of uncertain significance. No deletion/duplication in ASPM was identified in the patients analyzed. A wide ethnic distribution was observed, including the first reported patients with ASPM-related MCPH of Hispanic descent. Clinical information was collected for 26 of the ASPM-positive patients and 41 of the ASPM-negative patients. As more individuals are identified with MCPH, we anticipate that we will continue to identify ASPM mutation-positive patients from all ethnic origins supporting the occurrence of this genetic condition beyond that of consanguineous families of certain ethnic populations.
Collapse
Affiliation(s)
- C A Tan
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | | | | | | | | | | | | |
Collapse
|
71
|
Reynolds JJ, Stewart GS. A single strand that links multiple neuropathologies in human disease. ACTA ACUST UNITED AC 2013; 136:14-27. [PMID: 23365091 DOI: 10.1093/brain/aws310] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
The development of the human central nervous system is a complex process involving highly coordinated periods of neuronal proliferation, migration and differentiation. Disruptions in these neurodevelopmental processes can result in microcephaly, a neuropathological disorder characterized by a reduction in skull circumference and total brain volume, whereas a failure to maintain neuronal health in the adult brain can lead to progressive neurodegeneration. Defects in the cellular pathways that detect and repair DNA damage are a common cause of both these neuropathologies and are associated with a growing number of hereditary human disorders. In particular, defects in the repair of DNA single strand breaks, one of the most commonly occurring types of DNA lesion, have been associated with three neuropathological diseases: ataxia oculomotor apraxia 1, spinocerebellar ataxia with neuronal neuropathy 1 and microcephaly, early-onset, intractable seizures and developmental delay. A striking similarity between these three human diseases is that they are all caused by mutations in DNA end processing factors, suggesting that a particularly crucial stage of DNA single strand break repair is the repair of breaks with 'damaged' termini. Additionally all three disorders lack any extraneurological symptoms, such as immunodeficiency and cancer predisposition, which are typically found in other human diseases associated with defective DNA repair. However despite these similarities, two of these disorders present with progressive cerebellar degeneration, whereas the third presents with severe microcephaly. This review discusses the molecular defects behind these disorders and presents several hypotheses based on current literature on a number of important questions, in particular, how do mutations in different end processing factors within the same DNA repair pathway lead to such different neuropathologies?
Collapse
Affiliation(s)
- John J Reynolds
- School of Cancer Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | | |
Collapse
|
72
|
Zollikofer CPE, De León MSP. Pandora's growing box: Inferring the evolution and development of hominin brains from endocasts. Evol Anthropol 2013; 22:20-33. [DOI: 10.1002/evan.21333] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
73
|
Targeted disruption in mice of a neural stem cell-maintaining, KRAB-Zn finger-encoding gene that has rapidly evolved in the human lineage. PLoS One 2012; 7:e47481. [PMID: 23071813 PMCID: PMC3468564 DOI: 10.1371/journal.pone.0047481] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 09/17/2012] [Indexed: 11/19/2022] Open
Abstract
Understanding the genetic basis of the physical and behavioral traits that separate humans from other primates is a challenging but intriguing topic. The adaptive functions of the expansion and/or reduction in human brain size have long been explored. From a brain transcriptome project we have identified a KRAB-Zn finger protein-encoding gene (M003-A06) that has rapidly evolved since the human-chimpanzee separation. Quantitative RT-PCR analysis of different human tissues indicates that M003-A06 expression is enriched in the human fetal brain in addition to the fetal heart. Furthermore, analysis with use of immunofluorescence staining, neurosphere culturing and Western blotting indicates that the mouse ortholog of M003-A06, Zfp568, is expressed mainly in the embryonic stem (ES) cells and fetal as well as adult neural stem cells (NSCs). Conditional gene knockout experiments in mice demonstrates that Zfp568 is both an NSC maintaining- and a brain size-regulating gene. Significantly, molecular genetic analyses show that human M003-A06 consists of 2 equilibrated allelic types, H and C, one of which (H) is human-specific. Combined contemporary genotyping and database mining have revealed interesting genetic associations between the different genotypes of M003-A06 and the human head sizes. We propose that M003-A06 is likely one of the genes contributing to the uniqueness of the human brain in comparison to other higher primates.
Collapse
|
74
|
Peyre E, Morin X. An oblique view on the role of spindle orientation in vertebrate neurogenesis. Dev Growth Differ 2012; 54:287-305. [PMID: 22524602 DOI: 10.1111/j.1440-169x.2012.01350.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Neurogenesis is a dynamic process that produces a diverse number of glial and neural cell types from a limited number of neural stem cells throughout development and into adulthood. After an initial period of amplification through symmetric division, neural stem cells rely on asymmetric modes of division to self-renew while producing more committed progeny. Understanding the molecular mechanisms regulating the choice between symmetric and asymmetric modes of division is essential to understand human brain development and pathologies, and to explain the increasing cortical complexity observed in evolution. A popular model states the existence of a causal relationship between the orientation of the axis of division of stem cells and the fate of their progeny in many different tissues, but the validity of the model in neural stem cells is not clear. In this review, we briefly present the diversity of neural stem cells and intermediate progenitors in the developing central nervous system. We then draw a historic overview of the assumed causal relationship between spindle orientation and fate determination. We show how this prompted a search for regulators of spindle orientation, and present the current state of knowledge on the mechanism. Finally, we review data on the effect of defective spindle orientation and try to integrate conflicting observations by presenting alternative mechanisms that may regulate the choice between symmetric and asymmetric outcomes.
Collapse
Affiliation(s)
- Elise Peyre
- Institut de Biologie du Développement de Marseille-Luminy, CNRS UMR, 6216, case 907, Parc scientifique de Luminy, 13288, Marseille Cedex 9, France
| | | |
Collapse
|
75
|
Gavvovidis I, Rost I, Trimborn M, Kaiser FJ, Purps J, Wiek C, Hanenberg H, Neitzel H, Schindler D. A novel MCPH1 isoform complements the defective chromosome condensation of human MCPH1-deficient cells. PLoS One 2012; 7:e40387. [PMID: 22952573 PMCID: PMC3431399 DOI: 10.1371/journal.pone.0040387] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Accepted: 06/08/2012] [Indexed: 12/30/2022] Open
Abstract
Biallelic mutations in MCPH1 cause primary microcephaly (MCPH) with the cellular phenotype of defective chromosome condensation. MCPH1 encodes a multifunctional protein that notably is involved in brain development, regulation of chromosome condensation, and DNA damage response. In the present studies, we detected that MCPH1 encodes several distinct transcripts, including two major forms: full-length MCPH1 (MCPH1-FL) and a second transcript lacking the six 3′ exons (MCPH1Δe9–14). Both variants show comparable tissue-specific expression patterns, demonstrate nuclear localization that is mediated independently via separate NLS motifs, and are more abundant in certain fetal than adult organs. In addition, the expression of either isoform complements the chromosome condensation defect found in genetically MCPH1-deficient or MCPH1 siRNA-depleted cells, demonstrating a redundancy of both MCPH1 isoforms for the regulation of chromosome condensation. Strikingly however, both transcripts are regulated antagonistically during cell-cycle progression and there are functional differences between the isoforms with regard to the DNA damage response; MCPH1-FL localizes to phosphorylated H2AX repair foci following ionizing irradiation, while MCPH1Δe9–14 was evenly distributed in the nucleus. In summary, our results demonstrate here that MCPH1 encodes different isoforms that are differentially regulated at the transcript level and have different functions at the protein level.
Collapse
Affiliation(s)
- Ioannis Gavvovidis
- Department of Human Genetics, University of Würzburg, Würzburg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Abstract
The primary cilium is a microtubule-based structure protruded from the basal body analogous to the centriole. CPAP (centrosomal P4.1-associated protein) has previously been reported to be a cell cycle-regulated protein that controls centriole length. Mutations in CPAP cause primary microcephaly (MCPH) in humans. Here, using a cell-based system that we established to monitor cilia formation in neuronal CAD (Cath.a-differentiated) cells and hippocampal neurons, we found that CPAP is required for cilia biogenesis. Overexpression of wild-type CPAP promoted cilia formation and induced longer cilia. In contrast, an exogenously expressed CPAP-377EE mutant that lacks tubulin-dimer binding significantly inhibited cilia formation and caused cilia shortening. Furthermore, depletion of CPAP inhibited ciliogenesis and such effect was effectively rescued by expression of wild-type CPAP, but not by the CPAP-377EE mutant. Taken together, our results suggest that CPAP is a positive regulator of ciliogenesis whose intrinsic tubulin-dimer binding activity is required for cilia formation in neuronal cells.
Collapse
Affiliation(s)
- Kuo-Sheng Wu
- Program in Molecular Medicine, National Yang-Ming University and Academia Sinica , Nankang, Taipei 115 , Taiwan
| | | |
Collapse
|
77
|
Wong PCM, Chandrasekaran B, Zheng J. The derived allele of ASPM is associated with lexical tone perception. PLoS One 2012; 7:e34243. [PMID: 22529908 PMCID: PMC3328485 DOI: 10.1371/journal.pone.0034243] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 02/24/2012] [Indexed: 01/12/2023] Open
Abstract
The ASPM and MCPH1 genes have been implicated in the adaptive evolution of the human brain [Mekel-Bobrov N. et al., 2005. Ongoing adaptive evolution of ASPM, a brain size determinant in homo sapiens. Science 309; Evans P.D. et al., 2005. Microcephalin, a gene regulating brain size, continues to evolve adaptively in humans. Science 309]. Curiously, experimental attempts have failed to connect the implicated SNPs in these genes with higher-level brain functions. These results stand in contrast with a population-level study linking the population frequency of their alleles with the tendency to use lexical tones in a language [Dediu D., Ladd D.R., 2007. Linguistic tone is related to the population frequency of the adaptive haplogroups of two brain size genes, ASPM and microcephalin. Proc. Natl. Acad. Sci. U.S.A. 104]. In the present study, we found a significant correlation between the load of the derived alleles of ASPM and tone perception in a group of European Americans who did not speak a tone language. Moreover, preliminary results showed a significant correlation between ASPM load and hemodynamic responses to lexical tones in the auditory cortex, and such correlation remained after phonemic awareness, auditory working memory, and non-verbal IQ were controlled. As in previous studies, no significant correlation between ASPM and cognitive measures were found. MCPH1 did not correlate with any measures. These results suggest that the association between the recently derived allele of ASPM is likely to be specific and is tied to higher level brain functions in the temporal cortex related to human communication.
Collapse
Affiliation(s)
- Patrick C M Wong
- Roxelyn and Richard Pepper Department of Communication Sciences & Disorders, Northwestern University, Evanston, Illinois, United States of America.
| | | | | |
Collapse
|
78
|
Taal HR, Pourcain BS, Thiering E, Das S, Mook-Kanamori DO, Warrington NM, Kaakinen M, Kreiner-Møller E, Bradfield JP, Freathy RM, Geller F, Guxens M, Cousminer DL, Kerkhof M, Timpson NJ, Ikram MA, Beilin LJ, Bønnelykke K, Buxton JL, Charoen P, Chawes BLK, Eriksson J, Evans DM, Hofman A, Kemp JP, Kim CE, Klopp N, Lahti J, Lye SJ, McMahon G, Mentch FD, Müller M, O'Reilly PF, Prokopenko I, Rivadeneira F, Steegers EAP, Sunyer J, Tiesler C, Yaghootkar H, Breteler MMB, Debette S, Fornage M, Gudnason V, Launer LJ, van der Lugt A, Mosley TH, Seshadri S, Smith AV, Vernooij MW, Blakemore AI, Chiavacci RM, Feenstra B, Fernandez-Benet J, Grant SFA, Hartikainen AL, van der Heijden AJ, Iñiguez C, Lathrop M, McArdle WL, Mølgaard A, Newnham JP, Palmer LJ, Palotie A, Pouta A, Ring SM, Sovio U, Standl M, Uitterlinden AG, Wichmann HE, Vissing NH, DeCarli C, van Duijn CM, McCarthy MI, Koppelman GH, Estivill X, Hattersley AT, Melbye M, Bisgaard H, Pennell CE, Widen E, Hakonarson H, Smith GD, Heinrich J, Jarvelin MR, Jaddoe VWV. Common variants at 12q15 and 12q24 are associated with infant head circumference. Nat Genet 2012; 44:532-538. [PMID: 22504419 PMCID: PMC3773913 DOI: 10.1038/ng.2238] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 03/07/2012] [Indexed: 12/20/2022]
Abstract
To identify genetic variants associated with head circumference in infancy, we performed a meta-analysis of seven genome-wide association studies (GWAS) (N = 10,768 individuals of European ancestry enrolled in pregnancy and/or birth cohorts) and followed up three lead signals in six replication studies (combined N = 19,089). rs7980687 on chromosome 12q24 (P = 8.1 × 10(-9)) and rs1042725 on chromosome 12q15 (P = 2.8 × 10(-10)) were robustly associated with head circumference in infancy. Although these loci have previously been associated with adult height, their effects on infant head circumference were largely independent of height (P = 3.8 × 10(-7) for rs7980687 and P = 1.3 × 10(-7) for rs1042725 after adjustment for infant height). A third signal, rs11655470 on chromosome 17q21, showed suggestive evidence of association with head circumference (P = 3.9 × 10(-6)). SNPs correlated to the 17q21 signal have shown genome-wide association with adult intracranial volume, Parkinson's disease and other neurodegenerative diseases, indicating that a common genetic variant in this region might link early brain growth with neurological disease in later life.
Collapse
Affiliation(s)
- H Rob Taal
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Paediatrics, Erasmus Medical Center, Rotterdam, The Netherlands
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Beate St Pourcain
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Elisabeth Thiering
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Shikta Das
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
| | - Dennis O Mook-Kanamori
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Paediatrics, Erasmus Medical Center, Rotterdam, The Netherlands
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands
- Weill Cornell Medical College - Qatar, Doha, Qatar
| | - Nicole M Warrington
- School of Women's and Infants' Health, The University of Western Australia, Perth, Australia
- Samuel Lunenfeld Research Institute, University of Toronto, Toronto, Canada
| | - Marika Kaakinen
- Institute of Health Sciences, University of Oulu, Finland
- Biocenter Oulu, University of Oulu, Finland
| | - Eskil Kreiner-Møller
- Copenhagen Prospective Studies on Asthma in Childhood, University of Copenhagen, Copenhagen, Denmark
| | - Jonathan P Bradfield
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Rachel M Freathy
- Genetics of Complex Traits, Peninsula College of Medicine and Dentistry, University of Exeter, Exeter, UK
| | - Frank Geller
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Mònica Guxens
- Center for Research in Environmental Epidemiology (CREAL), Barcelona, Catalonia, Spain
- Hospital del Mar Research Institute (IMIM), Barcelona, Catalonia, Spain
- CIBER Epidemiologia y Salud Pública (CIBERESP), Barcelona, Catalonia, Spain
| | - Diana L Cousminer
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Marjan Kerkhof
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Nicholas J Timpson
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Radiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Lawrence J Beilin
- School of Medicine and Pharmacology, The University of Western Australia, Perth, Australia
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood, University of Copenhagen, Copenhagen, Denmark
| | - Jessica L Buxton
- Department of Genomics of Common Disease, School of Public Health, Imperial College London
| | - Pimphen Charoen
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Bo Lund Krogsgaard Chawes
- Copenhagen Prospective Studies on Asthma in Childhood, University of Copenhagen, Copenhagen, Denmark
| | - Johan Eriksson
- National Institute for Health and Welfare, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
- Folkhalsan Research Centre, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - David M Evans
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands
| | - John P Kemp
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Cecilia E Kim
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Norman Klopp
- Research Unit for Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Jari Lahti
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, Finland
| | - Stephen J Lye
- Samuel Lunenfeld Research Institute, University of Toronto, Toronto, Canada
| | - George McMahon
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Frank D Mentch
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Martina Müller
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Department of Medicine I, University Hospital Grosshadern, Ludwig-Maximilians-Universität, Munich, Germany
| | - Paul F O'Reilly
- Department of Epidemiology and Biostatistics, Imperial College London, W2 1PG London, UK
| | - Inga Prokopenko
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Eric A P Steegers
- Department of Obstetrics & Gynecology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jordi Sunyer
- Center for Research in Environmental Epidemiology (CREAL), Barcelona, Catalonia, Spain
- Hospital del Mar Research Institute (IMIM), Barcelona, Catalonia, Spain
- CIBER Epidemiologia y Salud Pública (CIBERESP), Barcelona, Catalonia, Spain
- Pompeu Fabra University (UPF), Barcelona, Catalonia, Spain
| | - Carla Tiesler
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Dr Von Hauner Children's Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Hanieh Yaghootkar
- Genetics of Complex Traits, Peninsula College of Medicine and Dentistry, University of Exeter, Exeter, UK
| | | | - Stephanie Debette
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Myriam Fornage
- Institute of Molecular Medicine, Human Genetics Center and Division of Epidemiology, School of Public Health, University of Texas, Houston Health Sciences Center, Houston, TX, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogus, Iceland
- University of Iceland, Reykjavik, Iceland
| | - Lenore J Launer
- Laboratory of Epidemiology, Demography and Biometry, National Institute on Aging, National Institute of Health, Bethesda, MD, USA
| | - Aad van der Lugt
- Department of Radiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Thomas H Mosley
- Department of Medicine (Geriatrics), University of Mississippi Medical Center, Jackson, MS, USA
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Albert V Smith
- Icelandic Heart Association, Kopavogus, Iceland
- University of Iceland, Reykjavik, Iceland
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Radiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Alexandra If Blakemore
- Department of Genomics of Common Disease, School of Public Health, Imperial College London
| | - Rosetta M Chiavacci
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Bjarke Feenstra
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Julio Fernandez-Benet
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, United Kingdom
| | - Struan F A Grant
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Department of Pediatrics, University of Pennsylvania, Philadelphia PA 19104, USA
| | - Anna-Liisa Hartikainen
- Institute of Clinical Medicine/Obstetrics and Gynecology, University of Oulu, Oulu, Finland
| | | | - Carmen Iñiguez
- CIBER Epidemiologia y Salud Pública (CIBERESP), Barcelona, Catalonia, Spain
- Division of Environment and Health, Center for Public Health Research-CSISP, Valencia, Spain
| | - Mark Lathrop
- Centre National de Génotypage, Evry, France
- Foundation Jean Dausset, CEPH, Paris, France
| | - Wendy L McArdle
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Anne Mølgaard
- Copenhagen Prospective Studies on Asthma in Childhood, University of Copenhagen, Copenhagen, Denmark
| | - John P Newnham
- School of Women's and Infants' Health, The University of Western Australia, Perth, Australia
| | - Lyle J Palmer
- Samuel Lunenfeld Research Institute, University of Toronto, Toronto, Canada
- Genetic Epidemiology and Biostatistics Platform, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Aarno Palotie
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Annneli Pouta
- National Institute for Health and Welfare, Oulu, Finland, Biocenter Oulu, University of Oulu, Finland
| | - Susan M Ring
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Ulla Sovio
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Marie Standl
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Andre G Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - H-Erich Wichmann
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
- Department of Medicine I, University Hospital Grosshadern, Ludwig-Maximilians-Universität, Munich, Germany
| | - Nadja Hawwa Vissing
- Copenhagen Prospective Studies on Asthma in Childhood, University of Copenhagen, Copenhagen, Denmark
| | - Charles DeCarli
- Department of Neurology and Center for Neuroscience, University of California at Davis, Sacramento, CA, USA
| | | | - Mark I McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Gerard H Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Xavier Estivill
- CIBER Epidemiologia y Salud Pública (CIBERESP), Barcelona, Catalonia, Spain
- Pompeu Fabra University (UPF), Barcelona, Catalonia, Spain
- Genes and Disease Program, Center for Genomic Regulation (CRG-UPF), Barcelona, Catalonia, Spain
| | - Andrew T Hattersley
- Peninsula NIHR Clinical Research Facility, Peninsula College of Medicine and Dentistry, University of Exeter, Exeter, UK
| | - Mads Melbye
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Hans Bisgaard
- Copenhagen Prospective Studies on Asthma in Childhood, University of Copenhagen, Copenhagen, Denmark
| | - Craig E Pennell
- School of Women's and Infants' Health, The University of Western Australia, Perth, Australia
| | - Elisabeth Widen
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Hakon Hakonarson
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Department of Pediatrics, University of Pennsylvania, Philadelphia PA 19104, USA
| | - George Davey Smith
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Joachim Heinrich
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Marjo-Riitta Jarvelin
- Institute of Health Sciences, University of Oulu, Finland
- National Institute for Health and Welfare, Oulu, Finland, Biocenter Oulu, University of Oulu, Finland
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, MRC Health Protection Agency (HPA) Centre for Environment and Health, Imperial College London
| | - Vincent W V Jaddoe
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Paediatrics, Erasmus Medical Center, Rotterdam, The Netherlands
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
79
|
|
80
|
Dediu D. Are languages really independent from genes? If not, what would a genetic bias affecting language diversity look like? Hum Biol 2011; 83:279-96. [PMID: 21615290 DOI: 10.3378/027.083.0208] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
It is generally accepted that the relationship between human genes and language is very complex and multifaceted. This has its roots in the “regular” complexity governing the interplay among genes and between genes and environment for most phenotypes, but with the added layer of supraontogenetic and supra-individual processes defining culture. At the coarsest level, focusing on the species, it is clear that human-specific--but not necessarily faculty-specific--genetic factors subtend our capacity for language and a currently very productive research program is aiming at uncovering them. At the other end of the spectrum, it is uncontroversial that individual-level variations in different aspects related to speech and language have an important genetic component and their discovery and detailed characterization have already started to revolutionize the way we think about human nature. However, at the intermediate, glossogenetic/population level, the relationship becomes controversial, partly due to deeply ingrained beliefs about language acquisition and universality and partly because of confusions with a different type of gene-languages correlation due to shared history. Nevertheless, conceptual, mathematical and computational models--and, recently, experimental evidence from artificial languages and songbirds--have repeatedly shown that genetic biases affecting the acquisition or processing of aspects of language and speech can be amplified by population-level intergenerational cultural processes and made manifest either as fixed “universal” properties of language or as structured linguistic diversity. Here, I review several such models as well as the recently proposed case of a causal relationship between the distribution of tone languages and two genes related to brain growth and development, ASPM and Microcephalin, and I discuss the relevance of such genetic biasing for language evolution, change, and diversity.
Collapse
Affiliation(s)
- Dan Dediu
- Max Planck Institute for Psycho linguistics, Wundtlaan 1, 6525 XD Nijmegen, the Netherlands.
| |
Collapse
|
81
|
Interkinetic nuclear movement in the ventricular zone of the cortex. J Mol Neurosci 2011; 46:516-26. [PMID: 21881827 DOI: 10.1007/s12031-011-9633-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2011] [Accepted: 08/17/2011] [Indexed: 10/17/2022]
Abstract
The nuclei of neuroepithelial cells move along the apicobasal axis in synchronization with their cell cycle status. This motility is known as interkinetic nuclear movement. We discuss here the importance of cytoskeleton organization, the centrosome, molecular motors, cell polarity proteins, and their regulators in controlling and maintaining this typical behavior. Furthermore, due to the tight linkage between cell proliferation, cell cycle, and nuclear motility, we speculate that interkinetic nuclear movement is likely to be affected in the pathophysiology of microcephaly, where the brain size is markedly reduced.
Collapse
|
82
|
Mahmood S, Ahmad W, Hassan MJ. Autosomal Recessive Primary Microcephaly (MCPH): clinical manifestations, genetic heterogeneity and mutation continuum. Orphanet J Rare Dis 2011; 6:39. [PMID: 21668957 PMCID: PMC3123551 DOI: 10.1186/1750-1172-6-39] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Accepted: 06/13/2011] [Indexed: 12/21/2022] Open
Abstract
Autosomal Recessive Primary Microcephaly (MCPH) is a rare disorder of neurogenic mitosis characterized by reduced head circumference at birth with variable degree of mental retardation. In MCPH patients, brain size reduced to almost one-third of its original volume due to reduced number of generated cerebral cortical neurons during embryonic neurogensis. So far, seven genetic loci (MCPH1-7) for this condition have been mapped with seven corresponding genes (MCPH1, WDR62, CDK5RAP2, CEP152, ASPM, CENPJ, and STIL) identified from different world populations. Contribution of ASPM and WDR62 gene mutations in MCPH World wide is more than 50%. By and large, primary microcephaly patients are phenotypically indistinguishable, however, recent studies in patients with mutations in MCPH1, WDR62 and ASPM genes showed a broader clinical and/or cellular phenotype. It has been proposed that mutations in MCPH genes can cause the disease phenotype by disturbing: 1) orientation of mitotic spindles, 2) chromosome condensation mechanism during embryonic neurogenesis, 3) DNA damage-response signaling, 4) transcriptional regulations and microtubule dynamics, 5) certain unknown centrosomal mechanisms that control the number of neurons generated by neural precursor cells. Recent discoveries of mammalian models for MCPH have open up horizons for researchers to add more knowledge regarding the etiology and pathophysiology of MCPH. High incidence of MCPH in Pakistani population reflects the most probable involvement of consanguinity. Genetic counseling and clinical management through carrier detection/prenatal diagnosis in MCPH families can help reducing the incidence of this autosomal recessive disorder.
Collapse
Affiliation(s)
- Saqib Mahmood
- Department of Human Genetics and Molecular Biology, University of Health Sciences, Khayaban-e-Jamia Punjab, Lahore, 54600, Pakistan
| | | | | |
Collapse
|
83
|
McGowen MR, Montgomery SH, Clark C, Gatesy J. Phylogeny and adaptive evolution of the brain-development gene microcephalin (MCPH1) in cetaceans. BMC Evol Biol 2011; 11:98. [PMID: 21492470 PMCID: PMC3101173 DOI: 10.1186/1471-2148-11-98] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Accepted: 04/14/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Representatives of Cetacea have the greatest absolute brain size among animals, and the largest relative brain size aside from humans. Despite this, genes implicated in the evolution of large brain size in primates have yet to be surveyed in cetaceans. RESULTS We sequenced ~1240 basepairs of the brain development gene microcephalin (MCPH1) in 38 cetacean species. Alignments of these data and a published complete sequence from Tursiops truncatus with primate MCPH1 were utilized in phylogenetic analyses and to estimate ω (rate of nonsynonymous substitution/rate of synonymous substitution) using site and branch models of molecular evolution. We also tested the hypothesis that selection on MCPH1 was correlated with brain size in cetaceans using a continuous regression analysis that accounted for phylogenetic history. Our analyses revealed widespread signals of adaptive evolution in the MCPH1 of Cetacea and in other subclades of Mammalia, however, there was not a significant positive association between ω and brain size within Cetacea. CONCLUSION In conjunction with a recent study of Primates, we find no evidence to support an association between MCPH1 evolution and the evolution of brain size in highly encephalized mammalian species. Our finding of significant positive selection in MCPH1 may be linked to other functions of the gene.
Collapse
Affiliation(s)
- Michael R McGowen
- Department of Biology, University of California, Riverside, 92521, USA.
| | | | | | | |
Collapse
|
84
|
Kim T, Park JSY, Lee P, Jin Y, Fu SB, Rosales JL, Lee KY. Novel alternatively spliced variant form of human CDK5RAP2. Cell Cycle 2011; 10:1010-2. [PMID: 21346412 DOI: 10.4161/cc.10.6.15121] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
85
|
Abstract
In this review, I describe how evolutionary genomics is uniquely suited to spearhead advances in understanding human disease risk, owing to the privileged position of genes as fundamental causes of phenotypic variation, and the ability of population genetic and phylogenetic methods to robustly infer processes of natural selection, drift, and mutation from genetic variation at the levels of family, population, species, and clade. I first provide an overview of models for the origins and maintenance of genetically based disease risk in humans. I then discuss how analyses of genetic disease risk can be dovetailed with studies of positive and balancing selection, to evaluate the degree to which the 'genes that make us human' also represent the genes that mediate risk of polygenic disease. Finally, I present four basic principles for the nascent field of human evolutionary medical genomics, each of which represents a process that is nonintuitive from a proximate perspective. Joint consideration of these principles compels novel forms of interdisciplinary analyses, most notably studies that (i) analyze tradeoffs at the level of molecular genetics, and (ii) identify genetic variants that are derived in the human lineage or in specific populations, and then compare individuals with derived versus ancestral alleles.
Collapse
Affiliation(s)
- Bernard J Crespi
- Department of Biosciences, Simon Fraser University Burnaby, BC, Canada
| |
Collapse
|
86
|
p21-Activated kinases 1 and 3 control brain size through coordinating neuronal complexity and synaptic properties. Mol Cell Biol 2010; 31:388-403. [PMID: 21115725 DOI: 10.1128/mcb.00969-10] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The molecular mechanisms that coordinate postnatal brain enlargement, synaptic properties, and cognition remain an enigma. Here, we demonstrate that neuronal complexity controlled by p21-activated kinases (PAKs) is a key determinant for postnatal brain enlargement and synaptic properties. We showed that double-knockout (DK) mice lacking both PAK1 and PAK3 were born healthy, with normal brain size and structure, but severely impaired in postnatal brain growth, resulting in a dramatic reduction in brain volume. Remarkably, the reduced brain size was accompanied by minimal changes in total cell count, due to a significant increase in cell density. However, the DK neurons have smaller soma, markedly simplified dendritic arbors/axons, and reduced synapse density. Surprisingly, the DK mice had elevated basal synaptic responses due to enhanced individual synaptic potency but were severely impaired in bidirectional synaptic plasticity. The actions of PAK1 and PAK3 are possibly mediated by cofilin-dependent actin regulation, because the activity of cofilin and the properties of actin filaments were altered in the DK mice. These results reveal an essential in vivo role of PAK1 and PAK3 in coordinating neuronal complexity and synaptic properties and highlight the critical importance of dendrite/axon growth in dictating postnatal brain growth and attainment of normal brain size and function.
Collapse
|
87
|
Dixon-Salazar TJ, Gleeson JG. Genetic regulation of human brain development: lessons from Mendelian diseases. Ann N Y Acad Sci 2010; 1214:156-67. [PMID: 21062301 DOI: 10.1111/j.1749-6632.2010.05819.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
One of the fundamental goals in human genetics is to link gene function to phenotype, yet the function of the majority of the genes in the human body is still poorly understood. This is especially true for the developing human brain. The study of human phenotypes that result from inherited, mutated alleles is the most direct evidence for the requirement of a gene in human physiology. Thus, the study of Mendelian central nervous system (CNS) diseases can be an extremely powerful approach to elucidate such phenotypic/genotypic links and to increase our understanding of the key components required for development of the human brain. In this review, we highlight examples of how the study of inherited neurodevelopmental disorders contributes to our knowledge of both the "normal" and diseased human brain, as well as elaborate on the future of this type of research. Mendelian disease research has been, and will continue to be, key to understanding the molecular mechanisms that underlie human brain function, and will ultimately form a basis for the design of intelligent, mechanism-specific treatments for nervous system disorders.
Collapse
Affiliation(s)
- Tracy J Dixon-Salazar
- Departments of Neurosciences and Pediatrics, Institute for Genomic Medicine, Howard Hughes Medical Institute, University of California, San Diego, La Jolla, California, USA
| | | |
Collapse
|
88
|
Montgomery SH, Capellini I, Venditti C, Barton RA, Mundy NI. Adaptive evolution of four microcephaly genes and the evolution of brain size in anthropoid primates. Mol Biol Evol 2010; 28:625-38. [PMID: 20961963 DOI: 10.1093/molbev/msq237] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The anatomical basis and adaptive function of the expansion in primate brain size have long been studied; however, we are only beginning to understand the genetic basis of these evolutionary changes. Genes linked to human primary microcephaly have received much attention as they have accelerated evolutionary rates along lineages leading to humans. However, these studies focus narrowly on apes, and the link between microcephaly gene evolution and brain evolution is disputed. We analyzed the molecular evolution of four genes associated with microcephaly (ASPM, CDK5RAP2, CENPJ, MCPH1) across 21 species representing all major clades of anthropoid primates. Contrary to prevailing assumptions, positive selection was not limited to or intensified along the lineage leading to humans. In fact we show that all four loci were subject to positive selection across the anthropoid primate phylogeny. We developed clearly defined hypotheses to explicitly test if selection on these loci was associated with the evolution of brain size. We found positive relationships between both CDK5RAP2 and ASPM and neonatal brain mass and somewhat weaker relationships between these genes and adult brain size. In contrast, there is no evidence linking CENPJ and MCPH1 to brain size evolution. The stronger association of ASPM and CDK5RAP2 evolution with neonatal brain size than with adult brain size is consistent with these loci having a direct effect on prenatal neuronal proliferation. These results suggest that primate brain size may have at least a partially conserved genetic basis. Our results contradict a previous study that linked adaptive evolution of ASPM to changes in relative cortex size; however, our analysis indicates that this conclusion is not robust. Our finding that the coding regions of two widely expressed loci has experienced pervasive positive selection in relation to a complex, quantitative developmental phenotype provides a notable counterexample to the commonly asserted hypothesis that cis-regulatory regions play a dominant role in phenotypic evolution.
Collapse
|
89
|
Stoler-Poria S, Lev D, Schweiger A, Lerman-Sagie T, Malinger G. Developmental outcome of isolated fetal microcephaly. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2010; 36:154-158. [PMID: 20069548 DOI: 10.1002/uog.7556] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
OBJECTIVE To assess the neurodevelopmental outcome of children with prenatally diagnosed isolated microcephaly defined as head circumference more than 2 SD below the gestational mean. METHODS Children aged 2-6 years who had been diagnosed in utero as having head circumference measurements more than 2 SD below the gestational mean were compared with normocephalic children, utilizing a standard neuropsychological battery. Comparisons were also made within the study group. RESULTS HCs were between -2 and -4.8 SD, only one fetus having a HC below -3 SD. Children with a prenatal head circumference between 2 SD and 3 SD below the gestational mean did not differ significantly from the control group regarding cognitive, language and motor functioning. However, they exhibited more behavioral-emotional problems. There were no significant differences between children who had suffered from intrauterine growth restriction and those who had not, or between those with familial and those with non-familial microcephaly. Nevertheless, linear regression analysis showed that head circumference in utero helps predict cognitive functioning later in life. CONCLUSIONS Prenatally diagnosed head circumference between 2 SD and 3 SD below the gestational mean is not a risk factor for later abnormal neuropsychological development.
Collapse
Affiliation(s)
- S Stoler-Poria
- Department of Behavioral Sciences, Academic College of Tel Aviv, Tel Aviv, Israel
| | | | | | | | | |
Collapse
|
90
|
Mutations in centrosomal protein CEP152 in primary microcephaly families linked to MCPH4. Am J Hum Genet 2010; 87:40-51. [PMID: 20598275 DOI: 10.1016/j.ajhg.2010.06.003] [Citation(s) in RCA: 141] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2010] [Revised: 05/27/2010] [Accepted: 06/04/2010] [Indexed: 12/31/2022] Open
Abstract
Primary microcephaly is a rare condition in which brain size is substantially diminished without other syndromic abnormalities. Seven autosomal loci have been genetically mapped, and the underlying causal genes have been identified for MCPH1, MCPH3, MCPH5, MCPH6, and MCPH7 but not for MCPH2 or MCPH4. The known genes play roles in mitosis and cell division. We ascertained three families from an Eastern Canadian subpopulation, each with one microcephalic child. Homozygosity analysis in two families using genome-wide dense SNP genotyping supported linkage to the published MCPH4 locus on chromosome 15q21.1. Sequencing of coding exons of candidate genes in the interval identified a nonconservative amino acid change in a highly conserved residue of the centrosomal protein CEP152. The affected children in these two families were both homozygous for this missense variant. The third affected child was compound heterozygous for the missense mutation plus a second, premature-termination mutation truncating a third of the protein and preventing its localization to centrosomes in transfected cells. CEP152 is the putative mammalian ortholog of Drosphila asterless, mutations in which affect mitosis in the fly. Published data from zebrafish are also consistent with a role of CEP152 in centrosome function. By RT-PCR, CEP152 is expressed in the embryonic mouse brain, similar to other MCPH genes. Like some other MCPH genes, CEP152 shows signatures of positive selection in the human lineage. CEP152 is a strong candidate for the causal gene underlying MCPH4 and may be an important gene in the evolution of human brain size.
Collapse
|
91
|
Abstract
The role of microcephaly genes in normal variation in human brain size has been controversial. New studies show that a link does exist and imply sex-specificity in microcephaly gene action during neurogenesis.
Collapse
|
92
|
Skorobogatko YV, Deuso J, Adolf-Bryfogle J, Nowak MG, Gong Y, Lippa CF, Vosseller K. Human Alzheimer's disease synaptic O-GlcNAc site mapping and iTRAQ expression proteomics with ion trap mass spectrometry. Amino Acids 2010; 40:765-79. [PMID: 20563614 DOI: 10.1007/s00726-010-0645-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Accepted: 05/27/2010] [Indexed: 12/31/2022]
Abstract
Neuronal synaptic functional deficits are linked to impaired learning and memory in Alzheimer's disease (AD). We recently demonstrated that O-GlcNAc, a novel cytosolic and nuclear carbohydrate post-translational modification, is enriched at neuronal synapses and positively regulates synaptic plasticity linked to learning and memory in mice. Reduced levels of O-GlcNAc have been observed in AD, suggesting a possible link to deficits in synaptic plasticity. Using lectin enrichment and mass spectrometry, we mapped several human cortical synaptic O-GlcNAc modification sites. Overlap in patterns of O-GlcNAcation between mouse and human appears to be high, as previously mapped mouse synaptic O-GlcNAc sites in Bassoon, Piccolo, and tubulin polymerization promoting protein p25 were identified in human. Novel O-GlcNAc modification sites were identified on Mek2 and RPN13/ADRM1. Mek2 is a signaling component of the Erk 1/2 pathway involved in synaptic plasticity. RPN13 is a component of the proteasomal degradation pathway. The potential interplay of phosphorylation with mapped O-GlcNAc sites, and possible implication of those sites in synaptic plasticity in normal versus AD states is discussed. iTRAQ is a powerful differential isotopic quantitative approach in proteomics. Pulsed Q dissociation (PQD) is a recently introduced fragmentation strategy that enables detection of low mass iTRAQ reporter ions in ion trap mass spectrometry. We optimized LTQ ion trap settings for PQD-based iTRAQ quantitation and demonstrated its utility in O-GlcNAc site mapping. Using iTRAQ, abnormal synaptic expression levels of several proteins previously implicated in AD pathology were observed in addition to novel changes in synaptic specific protein expression including Synapsin II.
Collapse
Affiliation(s)
- Yuliya V Skorobogatko
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, USA
| | | | | | | | | | | | | |
Collapse
|
93
|
Kousar R, Nawaz H, Khurshid M, Ali G, Khan SU, Mir H, Ayub M, Wali A, Ali N, Jelani M, Basit S, Ahmad W, Ansar M. Mutation analysis of the ASPM gene in 18 Pakistani families with autosomal recessive primary microcephaly. J Child Neurol 2010; 25:715-20. [PMID: 19808985 DOI: 10.1177/0883073809346850] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Autosomal recessive primary microcephaly (MCPH) is a rare neurological disorder, in which the patients exhibit reduced occipital frontal head circumference (>3 standard deviations) and mild-to-severe mental retardation. Autosomal recessive primary microcephaly is genetically heterogeneous and 7 loci have been reported to date. Mutations in ASPM (abnormal spindle-like, microcephaly associated) gene are the most common cause of autosomal recessive primary microcephaly in the majority of the reported families. In the current investigation, we have located and studied 21 families with autosomal recessive primary microcephaly. Genotyping using polymorphic microsatellite markers linked to 7 autosomal recessive primary microcephaly loci revealed linkage of 18 families to the MCPH5 locus. Sequence analysis of the ASPM gene in 18 linked families detected 2 novel nonsense mutations (c.2101C>T/p.Q701X; c.9492T>G/p.Y3164X) in 2 families and 2 novel deletion mutations (c.6686delGAAA/p.R2229TfsX9; c.77delG/p.G26AfsX41) in 2 other families. Three previously described mutations (c.3978G>A/p.W1326X; c.1260delTCAAGTC/p.S420SfsX32; c.9159delA/p.K3053NfsX4) were also detected in 11 families. These identified mutations extended the body of evidence implicating the ASPM gene in the pathogenesis of human hereditary primary microcephaly.
Collapse
Affiliation(s)
- Rizwana Kousar
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Buchman JJ, Tseng HC, Zhou Y, Frank CL, Xie Z, Tsai LH. Cdk5rap2 interacts with pericentrin to maintain the neural progenitor pool in the developing neocortex. Neuron 2010; 66:386-402. [PMID: 20471352 DOI: 10.1016/j.neuron.2010.03.036] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2010] [Indexed: 01/19/2023]
Abstract
Primary autosomal-recessive microcephaly (MCPH) and Majewski osteodysplastic primordial dwarfism type II (MOPDII) are both genetic diseases that result in decreased brain size at birth. MCPH is thought to arise from alterations in the size of the neural progenitor pool, but the cause of this defect has not been thoroughly explored. We find that one of the genes associated with MCPH, Cdk5rap2, is highly expressed in the neural progenitor pool and that its loss results in a depletion of apical progenitors and increased cell-cycle exit leading to premature neuronal differentiation. We link Cdk5rap2 function to the pericentriolar material protein pericentrin, loss of function of which is associated with MOPDII. Depletion of pericentrin in neural progenitors phenocopies effects of Cdk5rap2 knockdown and results in decreased recruitment of Cdk5rap2 to the centrosome. Our findings uncover a common mechanism, involving aberrations in the neurogenesis program, that may underlie the development of microcephaly in multiple diseases.
Collapse
Affiliation(s)
- Joshua J Buchman
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Building 46, Room 4235A, Cambridge, MA 02139, USA
| | | | | | | | | | | |
Collapse
|
95
|
Szabó N, Pap C, Kóbor J, Svékus A, Túri S, Sztriha L. Primary microcephaly in Hungary: epidemiology and clinical features. Acta Paediatr 2010; 99:690-693. [PMID: 20064139 DOI: 10.1111/j.1651-2227.2009.01666.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIM To describe the population-based epidemiological characteristics and clinical features of primary microcephaly in Hungary. METHODS A retrospective survey of patients born with microcephaly in a region (Dél-Alföld - South Great Plain) in Hungary between July 1, 1992 and June 30, 2006 was performed. Patients with microcephaly and without any environmental or obstetric risk factors and/or dysmorphism (primary microcephaly) were included in the study. The birth prevalence of primary microcephaly per 10,000 live births was calculated. RESULTS Ten patients (8 girls and 2 boys) were found with primary microcephaly among 185,486 live births, which corresponds to a birth prevalence of 0.54 per 10,000 live births (95% confidence interval: 0.20-0.87). Developmental delay and intellectual disability were the main clinical features. Dyskinesia was seen in one and epilepsy was diagnosed in two patients. The MRI revealed simplified gyral pattern in all patients. CONCLUSION Primary microcephaly is a very rare brain malformation, although the birth prevalence found in this survey is slightly higher than the few figures published earlier. As more and more genes and mutations responsible for primary microcephaly are discovered, the ascertainment of these rare cases is mandatory to provide the parents with genetic counselling.
Collapse
Affiliation(s)
- Nóra Szabó
- Department of Paediatrics, Faculty of Medicine, University of Szeged, Szeged and Pándy Kálmán County Hospital, Gyula, Hungary
| | - Csenge Pap
- Department of Paediatrics, Faculty of Medicine, University of Szeged, Szeged and Pándy Kálmán County Hospital, Gyula, Hungary
| | - Jenő Kóbor
- Department of Paediatrics, Faculty of Medicine, University of Szeged, Szeged and Pándy Kálmán County Hospital, Gyula, Hungary
| | - András Svékus
- Department of Paediatrics, Faculty of Medicine, University of Szeged, Szeged and Pándy Kálmán County Hospital, Gyula, Hungary
| | - Sándor Túri
- Department of Paediatrics, Faculty of Medicine, University of Szeged, Szeged and Pándy Kálmán County Hospital, Gyula, Hungary
| | - László Sztriha
- Department of Paediatrics, Faculty of Medicine, University of Szeged, Szeged and Pándy Kálmán County Hospital, Gyula, Hungary
| |
Collapse
|
96
|
Holton NE, Franciscus RG, Nieves MA, Marshall SD, Reimer SB, Southard TE, Keller JC, Maddux SD. Sutural growth restriction and modern human facial evolution: an experimental study in a pig model. J Anat 2010; 216:48-61. [PMID: 19929910 PMCID: PMC2807975 DOI: 10.1111/j.1469-7580.2009.01162.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2009] [Indexed: 11/30/2022] Open
Abstract
Facial size reduction and facial retraction are key features that distinguish modern humans from archaic Homo. In order to more fully understand the emergence of modern human craniofacial form, it is necessary to understand the underlying evolutionary basis for these defining characteristics. Although it is well established that the cranial base exerts considerable influence on the evolutionary and ontogenetic development of facial form, less emphasis has been placed on developmental factors intrinsic to the facial skeleton proper. The present analysis was designed to assess anteroposterior facial reduction in a pig model and to examine the potential role that this dynamic has played in the evolution of modern human facial form. Ten female sibship cohorts, each consisting of three individuals, were allocated to one of three groups. In the experimental group (n = 10), microplates were affixed bilaterally across the zygomaticomaxillary and frontonasomaxillary sutures at 2 months of age. The sham group (n = 10) received only screw implantation and the controls (n = 10) underwent no surgery. Following 4 months of post-surgical growth, we assessed variation in facial form using linear measurements and principal components analysis of Procrustes scaled landmarks. There were no differences between the control and sham groups; however, the experimental group exhibited a highly significant reduction in facial projection and overall size. These changes were associated with significant differences in the infraorbital region of the experimental group including the presence of an infraorbital depression and an inferiorly and coronally oriented infraorbital plane in contrast to a flat, superiorly and sagittally infraorbital plane in the control and sham groups. These altered configurations are markedly similar to important additional facial features that differentiate modern humans from archaic Homo, and suggest that facial length restriction via rigid plate fixation is a potentially useful model to assess the developmental factors that underlie changing patterns in craniofacial form associated with the emergence of modern humans.
Collapse
Affiliation(s)
- Nathan E Holton
- Department of Orthodontics, University of Iowa, Iowa City, IA 52242, USA.
| | | | | | | | | | | | | | | |
Collapse
|
97
|
Crespi B, Summers K, Dorus S. Evolutionary genomics of human intellectual disability. Evol Appl 2010; 3:52-63. [PMID: 25567903 PMCID: PMC3352458 DOI: 10.1111/j.1752-4571.2009.00098.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Accepted: 07/28/2009] [Indexed: 01/28/2023] Open
Abstract
Previous studies have postulated that X-linked and autosomal genes underlying human intellectual disability may have also mediated the evolution of human cognition. We have conducted the first comprehensive assessment of the extent and patterns of positive Darwinian selection on intellectual disability genes in humans. We report three main findings. First, as noted in some previous reports, intellectual disability genes with primary functions in the central nervous system exhibit a significant concentration to the X chromosome. Second, there was no evidence for a higher incidence of recent positive selection on X-linked than autosomal intellectual disability genes, nor was there a higher incidence of selection on such genes overall, compared to sets of control genes. However, the X-linked intellectual disability genes inferred to be subject to recent positive selection were concentrated in the Rho GTP-ase pathway, a key signaling pathway in neural development and function. Third, among all intellectual disability genes, there was evidence for a higher incidence of recent positive selection on genes involved in DNA repair, but not for genes involved in other functions. These results provide evidence that alterations to genes in the Rho GTP-ase and DNA-repair pathways may play especially-important roles in the evolution of human cognition and vulnerability to genetically-based intellectual disability.
Collapse
Affiliation(s)
- Bernard Crespi
- Department of Biosciences, Simon Fraser UniversityBurnaby, BC, Canada
| | - Kyle Summers
- Department of Biology, East Carolina UniversityGreenville, NC, USA
| | - Steve Dorus
- Department of Biology and Biochemistry, University of BathBath, UK
| |
Collapse
|
98
|
Asymmetric centrosome inheritance maintains neural progenitors in the neocortex. Nature 2009; 461:947-55. [PMID: 19829375 PMCID: PMC2764320 DOI: 10.1038/nature08435] [Citation(s) in RCA: 360] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Accepted: 08/18/2009] [Indexed: 01/12/2023]
Abstract
Asymmetric divisions of radial glia progenitors produce self-renewing radial glia and differentiating cells simultaneously in the ventricular zone (VZ) of the developing neocortex. Whereas differentiating cells leave the VZ to constitute the future neocortex, renewing radial glia progenitors stay in the VZ for subsequent divisions. The differential behaviour of progenitors and their differentiating progeny is essential for neocortical development; however, the mechanisms that ensure these behavioural differences are unclear. Here we show that asymmetric centrosome inheritance regulates the differential behaviour of renewing progenitors and their differentiating progeny in the embryonic mouse neocortex. Centrosome duplication in dividing radial glia progenitors generates a pair of centrosomes with differently aged mother centrioles. During peak phases of neurogenesis, the centrosome retaining the old mother centriole stays in the VZ and is preferentially inherited by radial glia progenitors, whereas the centrosome containing the new mother centriole mostly leaves the VZ and is largely associated with differentiating cells. Removal of ninein, a mature centriole-specific protein, disrupts the asymmetric segregation and inheritance of the centrosome and causes premature depletion of progenitors from the VZ. These results indicate that preferential inheritance of the centrosome with the mature older mother centriole is required for maintaining radial glia progenitors in the developing mammalian neocortex.
Collapse
|
99
|
Zhao L, Jin C, Chu Y, Varghese C, Hua S, Yan F, Miao Y, Liu J, Mann D, Ding X, Zhang J, Wang Z, Dou Z, Yao X. Dimerization of CPAP orchestrates centrosome cohesion plasticity. J Biol Chem 2009; 285:2488-97. [PMID: 19889632 DOI: 10.1074/jbc.m109.042614] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Centrosome cohesion and segregation are accurately regulated to prevent an aberrant separation of duplicated centrosomes and to ensure the correct formation of bipolar spindles by a tight coupling with cell cycle machinery. CPAP is a centrosome protein with five coiled-coil domains and plays an important role in the control of brain size in autosomal recessive primary microcephaly. Previous studies showed that CPAP interacts with tubulin and controls centriole length. Here, we reported that CPAP forms a homodimer during interphase, and the fifth coiled-coil domain of CPAP is required for its dimerization. Moreover, this self-interaction is required for maintaining centrosome cohesion and preventing the centrosome from splitting before the G(2)/M phase. Our biochemical studies show that CPAP forms homodimers in vivo. In addition, both monomeric and dimeric CPAP are required for accurate cell division, suggesting that the temporal dynamics of CPAP homodimerization is tightly regulated during the cell cycle. Significantly, our results provide evidence that CPAP is phosphorylated during mitosis, and this phosphorylation releases its intermolecular interaction. Taken together, these results suggest that cell cycle-regulated phosphorylation orchestrates the dynamics of CPAP molecular interaction and centrosome splitting to ensure genomic stability in cell division.
Collapse
Affiliation(s)
- Lingli Zhao
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology and Hefei National Laboratory for Physical Sciences at Nanoscale, Hefei 230027, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Thornton GK, Woods CG. Primary microcephaly: do all roads lead to Rome? Trends Genet 2009; 25:501-10. [PMID: 19850369 PMCID: PMC2816178 DOI: 10.1016/j.tig.2009.09.011] [Citation(s) in RCA: 302] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Revised: 09/24/2009] [Accepted: 09/24/2009] [Indexed: 02/06/2023]
Abstract
The relatively large brain and expanded cerebral cortex of humans is unusual in the animal kingdom and is thought to have promoted our adaptability and success as a species. One approach for investigating neurogenesis is the study of autosomal recessive primary microcephaly (MCPH), in which prenatal brain growth is significantly reduced without an effect on brain structure. To date, eight MCPH loci and five genes have been identified. Unexpectedly, all MCPH proteins are ubiquitous and localise to centrosomes for at least part of the cell cycle. Here, we focus on recent functional studies of MCPH proteins that reveal the centrosome as a final integration point for many regulatory pathways affecting prenatal neurogenesis in mammals.
Collapse
Affiliation(s)
- Gemma K Thornton
- Department of Medical Genetics, Cambridge Institute for Medical Research, Wellcome/MRC Building, Addenbrookes Hospital, Cambridge, UK
| | | |
Collapse
|