101
|
Bouchelion A, Zhang Z, Li Y, Qian H, Mukherjee AB. Mice homozygous for c.451C>T mutation in Cln1 gene recapitulate INCL phenotype. Ann Clin Transl Neurol 2014; 1:1006-23. [PMID: 25574475 PMCID: PMC4284126 DOI: 10.1002/acn3.144] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Accepted: 10/16/2014] [Indexed: 11/21/2022] Open
Abstract
Objective Nonsense mutations account for 5–70% of all genetic disorders. In the United States, nonsense mutations in the CLN1/PPT1 gene underlie >40% of the patients with infantile neuronal ceroid lipofuscinosis (INCL), a devastating neurodegenerative lysosomal storage disease. We sought to generate a reliable mouse model of INCL carrying the most common Ppt1 nonsense mutation (c.451C>T) found in the United States patient population to provide a platform for evaluating nonsense suppressors in vivo. Methods We knocked-in c.451C>T nonsense mutation in the Ppt1 gene in C57 embryonic stem (ES) cells using a targeting vector in which LoxP flanked the Neo cassette, which was removed from targeted ES cells by electroporating Cre. Two independently targeted ES clones were injected into blastocysts to generate syngenic C57 knock-in mice, obviating the necessity for extensive backcrossing. Results Generation of Ppt1-KI mice was confirmed by DNA sequencing, which showed the presence of c.451C>T mutation in the Ppt1 gene. These mice are viable and fertile, although they developed spasticity (a “clasping” phenotype) at a median age of 6 months. Autofluorescent storage materials accumulated throughout the brain regions and in visceral organs. Electron microscopic analysis of the brain and the spleen showed granular osmiophilic deposits. Increased neuronal apoptosis was particularly evident in cerebral cortex and abnormal histopathological and electroretinographic (ERG) analyses attested striking retinal degeneration. Progressive deterioration of motor coordination and behavioral parameters continued until eventual death. Interpretation Our findings show that Ppt1-KI mice reliably recapitulate INCL phenotype providing a platform for testing the efficacy of existing and novel nonsense suppressors in vivo.
Collapse
Affiliation(s)
- Ashleigh Bouchelion
- Program on Developmental Endocrinology and Genetics, Section on Developmental Genetics, Eunice Kennedy-Shriver National Institute of Child Health and Human Development Bethesda, Maryland
| | - Zhongjian Zhang
- Program on Developmental Endocrinology and Genetics, Section on Developmental Genetics, Eunice Kennedy-Shriver National Institute of Child Health and Human Development Bethesda, Maryland
| | - Yichao Li
- Visual Function Core (HNW2-L), National Eye Institute, National Institutes of Health Bethesda, Maryland, 20892-1830
| | - Haohua Qian
- Visual Function Core (HNW2-L), National Eye Institute, National Institutes of Health Bethesda, Maryland, 20892-1830
| | - Anil B Mukherjee
- Program on Developmental Endocrinology and Genetics, Section on Developmental Genetics, Eunice Kennedy-Shriver National Institute of Child Health and Human Development Bethesda, Maryland
| |
Collapse
|
102
|
Jin M, Yamada M, Arai Y, Nagai T, Hirotsune S. Arl3 and LC8 regulate dissociation of dynactin from dynein. Nat Commun 2014; 5:5295. [PMID: 25342295 DOI: 10.1038/ncomms6295] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 09/18/2014] [Indexed: 12/16/2022] Open
Abstract
Cytoplasmic dynein acts as a motor for the intracellular retrograde motility of vesicles and organelles along microtubules. However, the regulatory mechanism underlying release of dynactin bound cargoes from dynein motor remains largely unknown. Here we report that ADP-ribosylation factor-like 3 (Arl3) and dynein light chain LC8 induce dissociation of dynactin from dynein. Immunoprecipitation and microtubule pull-down assays revealed that Arl3(Q71L) and LC8 facilitated detachment of dynactin from dynein. We also demonstrated Arl3(Q71L) or LC8-mediated dynactin release from a dynein-dynactin complex through trace experiments using quantum dot (Qdot)-conjugated proteins. Furthermore, we disclosed interactions of Arl3 and LC8 with dynactin and dynein, respectively, by live-cell imaging. Finally, knockdown (KD) of Arl3 and LC8 by siRNA induced abnormal localizations of dynein, dynactin and related organelles. Our findings uncovered the surprising functional relevance of GTP-bound Arl3 and LC8 for the unloading regulation of dynactin-bound cargo from dynein motor.
Collapse
Affiliation(s)
- Mingyue Jin
- Department of Genetic Disease Research, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno, Osaka 545-8585, Japan
| | - Masami Yamada
- Department of Genetic Disease Research, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno, Osaka 545-8585, Japan
| | - Yoshiyuki Arai
- Department of Biomolecular Science and Engineering, Institute of Scientific and Industrial Research, Osaka University, Mihoga-oka 8-1, Osaka 567-0047, Japan
| | - Takeharu Nagai
- Department of Biomolecular Science and Engineering, Institute of Scientific and Industrial Research, Osaka University, Mihoga-oka 8-1, Osaka 567-0047, Japan
| | - Shinji Hirotsune
- Department of Genetic Disease Research, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno, Osaka 545-8585, Japan
| |
Collapse
|
103
|
Watrin F, Manent JB, Cardoso C, Represa A. Causes and consequences of gray matter heterotopia. CNS Neurosci Ther 2014; 21:112-22. [PMID: 25180909 DOI: 10.1111/cns.12322] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 07/30/2014] [Accepted: 08/06/2014] [Indexed: 12/17/2022] Open
Abstract
The objective of this article is to review the pathophysiological bases of gray matter heterotopia and to appreciate their involvement in brain cortical development and functional consequences, namely epilepsy. The development of the cerebral cortex results from complex sequential processes including cell proliferation, cell migration, cortical organization, and formation of neuronal networks. Disruption of these steps yields different types of cortical malformations including gray matter heterotopia, characterized by the ectopic position of neurons along the ventricular walls or in the deep white matter. Cortical malformations are major causes of epilepsy, being responsible for up to 40% of drug-resistant epilepsy, and the cognitive level of affected patients varies from normal to severely impaired. This review reports data from human patients and animal models highlighting the genetic causes for these disorders affecting not only neuronal migration but also the proliferation of cortical progenitors. Therefore, gray matter heterotopias should not be considered as solely due to an abnormal neuronal migration and classifying them as such may be too restrictive. The review will also summarize literature data indicating that besides ectopic neurons, neighbor cortical areas also play a consistent role in epileptogenesis, supporting the notion that plastic changes secondary to the initial malformation are instrumental in the pathophysiology of epilepsy in affected patients.
Collapse
Affiliation(s)
- Françoise Watrin
- INSERM, INMED, Marseille, France; Aix-Marseille University, UMR 901, Marseille, France
| | | | | | | |
Collapse
|
104
|
Tanabe Y, Shiota A, Kouroku-Murakami Y, Fujita-Jimbo E, Urase K, Takahashi K, Mezaki Y, Senoo H, Momoi T. Spatial and temporal expression of RA70/Scap2 in the developing neural tube. Neurosci Lett 2014; 576:1-5. [PMID: 24846415 DOI: 10.1016/j.neulet.2014.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 04/18/2014] [Accepted: 05/12/2014] [Indexed: 10/25/2022]
Abstract
Src kinase-associated phosphoprotein 2 (Ra70/scap2), which was originally isolated as a retinoic acid (RA)-induced gene, associates with molecules that modulate integrin-survival signals. Although RA is essential for vertebrate organogenesis in the posterior region, little is known about the biological role of RA70/Scap2 during development. In the present study, we demonstrate that Ra70/scap2 mRNA is temporally expressed during the RA-induced neuronal differentiation of P19 embryonic carcinoma cells. Homozygous knockout mice in which the Ra70/scap2 gene was replaced with LacZ exhibited embryonic lethality, while heterozygous mice displayed preferential expression of LacZ in posterior neural tissues, including the neural tube and hindbrain during development (E7.5-11.5), but not the forebrain. Ra70/scap2 was expressed in the ependymal layer and ventricular zone in the neural tube, where neuroepithelial cells and neuroblasts with proliferation capacity are localized, respectively. Thus, RA70/Scap2 may be necessary for RA-induced neuronal differentiation from the posterior neuroectoderm.
Collapse
Affiliation(s)
- Yuko Tanabe
- Center for Medical Science, International University of Health and Welfare, Kitakanemaru, Ohtawara, Tochigi, Japan
| | - Akira Shiota
- PhoenixBio, Ltd., Iwazo, Utsunomiya, Tochigi, Japan
| | - Yoriko Kouroku-Murakami
- Center for Medical Science, International University of Health and Welfare, Kitakanemaru, Ohtawara, Tochigi, Japan
| | - Eriko Fujita-Jimbo
- Center for Medical Science, International University of Health and Welfare, Kitakanemaru, Ohtawara, Tochigi, Japan; Department of Pediatrics, Jichi Medical University, Yakushiji, Shimotsukeshi, Tochigi, Japan
| | - Koko Urase
- Department of Biology, School of Medicine Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Kana Takahashi
- Department of Cell Biology and Morphology, Akita University Graduate School of Medicine, Hondo, Akita, Japan
| | - Yoshihiro Mezaki
- Department of Cell Biology and Morphology, Akita University Graduate School of Medicine, Hondo, Akita, Japan
| | - Haruki Senoo
- Department of Cell Biology and Morphology, Akita University Graduate School of Medicine, Hondo, Akita, Japan
| | - Takashi Momoi
- Center for Medical Science, International University of Health and Welfare, Kitakanemaru, Ohtawara, Tochigi, Japan.
| |
Collapse
|
105
|
Yu YR, You LR, Yan YT, Chen CM. Role of OVCA1/DPH1 in craniofacial abnormalities of Miller–Dieker syndrome. Hum Mol Genet 2014; 23:5579-96. [DOI: 10.1093/hmg/ddu273] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
106
|
Mutations in Eml1 lead to ectopic progenitors and neuronal heterotopia in mouse and human. Nat Neurosci 2014; 17:923-33. [DOI: 10.1038/nn.3729] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 04/29/2014] [Indexed: 12/17/2022]
|
107
|
Kiss A, Horvath P, Rothballer A, Kutay U, Csucs G. Nuclear motility in glioma cells reveals a cell-line dependent role of various cytoskeletal components. PLoS One 2014; 9:e93431. [PMID: 24691067 PMCID: PMC3972233 DOI: 10.1371/journal.pone.0093431] [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: 10/15/2013] [Accepted: 03/05/2014] [Indexed: 12/31/2022] Open
Abstract
Nuclear migration is a general term for the movement of the nucleus towards a specific site in the cell. These movements are involved in a number of fundamental biological processes, such as fertilization, cell division, and embryonic development. Despite of its importance, the mechanism of nuclear migration is still poorly understood in mammalian cells. In order to shed light on the mechanical processes underlying nuclear movements, we adapted a micro-patterning based assay. C6 rat and U87 human glioma cells seeded on fibronectin patterns - thereby forced into a bipolar morphology - displayed oscillatory movements of the nucleus or the whole cell, respectively. We found that both the actomyosin system and microtubules are involved in the nuclear/cellular movements of both cell lines, but their contributions are cell-/migration-type specific. Dynein activity was necessary for nuclear migration of C6 cells but active myosin-II was dispensable. On the other hand, coupled nuclear and cellular movements of U87 cells were driven by actomyosin contraction. We explain these cell-line dependent effects by the intrinsic differences in the overall mechanical tension due to the various cytoskeletal elements inside the cell. Our observations showed that the movements of the nucleus and the centrosome are strongly correlated and display large variation, indicating a tight but flexible coupling between them. The data also indicate that the forces responsible for nuclear movements are not acting directly via the centrosome. Based on our observations, we propose a new model for nuclear oscillations in C6 cells in which dynein and microtubule dynamics are the main drivers of nuclear movements. This mechanism is similar to the meiotic nuclear oscillations of Schizosaccharomyces pombe and may be evolutionary conserved.
Collapse
Affiliation(s)
- Alexa Kiss
- Institute of Biochemistry, ETH Zurich, Zurich, Switzerland; Light Microscopy and Screening Centre, ETH Zurich, Zurich, Switzerland
| | - Peter Horvath
- Institute of Biochemistry, ETH Zurich, Zurich, Switzerland; Light Microscopy and Screening Centre, ETH Zurich, Zurich, Switzerland
| | | | - Ulrike Kutay
- Institute of Biochemistry, ETH Zurich, Zurich, Switzerland
| | - Gabor Csucs
- Institute of Biochemistry, ETH Zurich, Zurich, Switzerland; Light Microscopy and Screening Centre, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
108
|
Moon HM, Wynshaw-Boris A. Cytoskeleton in action: lissencephaly, a neuronal migration disorder. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2014; 2:229-45. [PMID: 23495356 DOI: 10.1002/wdev.67] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
During neocortical development, the extensive migratory movements of neurons from their place of birth to their final location are essential for the coordinated wiring of synaptic circuits and proper neurological function. Failure or delay in neuronal migration causes severe abnormalities in cortical layering, which consequently results in human lissencephaly ('smooth brain'), a neuronal migration disorder. The brains of lissencephaly patients have less-convoluted gyri in the cerebral cortex with impaired cortical lamination of neurons. Since microtubule (MT) and actin-associated proteins play important functions in regulating the dynamics of MT and actin cytoskeletons during neuronal migration, genetic mutations or deletions of crucial genes involved in cytoskeletal processes lead to lissencephaly in human and neuronal migration defects in mouse. During neuronal migration, MT organization and transport are controlled by platelet-activating factor acetylhydrolase isoform 1b regulatory subunit 1 (PAFAH1B1, formerly known as LIS1, Lissencephaly-1), doublecortin (DCX), YWHAE, and tubulin. Actin stress fibers are modulated by PAFAH1B1 (LIS1), DCX, RELN, and VLDLR (very low-density lipoprotein receptor)/LRP8 (low-density lipoprotein-related receptor 8, formerly known as APOER2). There are several important levels of crosstalk between these two cytoskeletal systems to establish accurate cortical patterning in development. The recent understanding of the protein networks that govern neuronal migration by regulating cytoskeletal dynamics, from human and mouse genetics as well as molecular and cellular analyses, provides new insights on neuronal migration disorders and may help us devise novel therapeutic strategies for such brain malformations.
Collapse
|
109
|
Belvindrah R, Nosten-Bertrand M, Francis F. Neuronal migration and its disorders affecting the CA3 region. Front Cell Neurosci 2014; 8:63. [PMID: 24624057 PMCID: PMC3941003 DOI: 10.3389/fncel.2014.00063] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 02/13/2014] [Indexed: 11/15/2022] Open
Abstract
In this review, we focus on CA3 neuronal migration disorders in the rodent. We begin by introducing the main steps of hippocampal development, and we summarize characteristic hippocampal malformations in human. We then describe various mouse mutants showing structural hippocampal defects. Notably, genes identified in human cortical neuronal migration disorders consistently give rise to a CA3 phenotype when mutated in the mouse. We successively describe their molecular, physiological and behavioral phenotypes that together contribute to a better understanding of CA3-dependent functions. We finally discuss potential factors underlying the CA3 vulnerability revealed by these mouse mutants and that may also contribute to other human neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Richard Belvindrah
- INSERM UMR-S 839 Paris, France ; Sorbonne Universités, Université Pierre et Marie Curie, Univ Paris 06 Paris, France ; Institut du Fer à Moulin Paris, France
| | - Marika Nosten-Bertrand
- INSERM UMR-S 839 Paris, France ; Sorbonne Universités, Université Pierre et Marie Curie, Univ Paris 06 Paris, France ; Institut du Fer à Moulin Paris, France
| | - Fiona Francis
- INSERM UMR-S 839 Paris, France ; Sorbonne Universités, Université Pierre et Marie Curie, Univ Paris 06 Paris, France ; Institut du Fer à Moulin Paris, France
| |
Collapse
|
110
|
Zimdahl B, Ito T, Blevins A, Bajaj J, Konuma T, Weeks J, Koechlein CS, Kwon HY, Arami O, Rizzieri D, Broome HE, Chuah C, Oehler VG, Sasik R, Hardiman G, Reya T. Lis1 regulates asymmetric division in hematopoietic stem cells and in leukemia. Nat Genet 2014; 46:245-52. [PMID: 24487275 PMCID: PMC4267534 DOI: 10.1038/ng.2889] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 01/09/2014] [Indexed: 01/08/2023]
Abstract
Cell fate can be controlled through asymmetric division and segregation of protein determinants, but the regulation of this process in the hematopoietic system is poorly understood. Here we show that the dynein-binding protein Lis1 is critically required for hematopoietic stem cell function and leukemogenesis. Conditional deletion of Lis1 (also known as Pafah1b1) in the hematopoietic system led to a severe bloodless phenotype, depletion of the stem cell pool and embryonic lethality. Further, real-time imaging revealed that loss of Lis1 caused defects in spindle positioning and inheritance of cell fate determinants, triggering accelerated differentiation. Finally, deletion of Lis1 blocked the propagation of myeloid leukemia and led to a marked improvement in survival, suggesting that Lis1 is also required for oncogenic growth. These data identify a key role for Lis1 in hematopoietic stem cells and mark its directed control of asymmetric division as a critical regulator of normal and malignant hematopoietic development.
Collapse
Affiliation(s)
- Bryan Zimdahl
- Department of Pharmacology, University of California San Diego School of Medicine, La Jolla, CA, 92093
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710
| | - Takahiro Ito
- Department of Pharmacology, University of California San Diego School of Medicine, La Jolla, CA, 92093
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093
| | - Allen Blevins
- Department of Pharmacology, University of California San Diego School of Medicine, La Jolla, CA, 92093
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093
| | - Jeevisha Bajaj
- Department of Pharmacology, University of California San Diego School of Medicine, La Jolla, CA, 92093
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093
| | - Takaaki Konuma
- Department of Pharmacology, University of California San Diego School of Medicine, La Jolla, CA, 92093
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093
| | - Joi Weeks
- Department of Pharmacology, University of California San Diego School of Medicine, La Jolla, CA, 92093
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093
| | - Claire S. Koechlein
- Department of Pharmacology, University of California San Diego School of Medicine, La Jolla, CA, 92093
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093
| | - Hyog Young Kwon
- Department of Pharmacology, University of California San Diego School of Medicine, La Jolla, CA, 92093
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093
| | - Omead Arami
- Department of Pharmacology, University of California San Diego School of Medicine, La Jolla, CA, 92093
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093
| | - David Rizzieri
- Division of Cell Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, 27710
| | - H. Elizabeth Broome
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, CA, 92093
- Moores Cancer Center, University of California San Diego School of Medicine, La Jolla, CA, 92093
| | - Charles Chuah
- Department of Haematology, Singapore General Hospital, Singapore
| | - Vivian G. Oehler
- Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109
| | - Roman Sasik
- Department of Medicine, University of California San Diego School of Medicine, La Jolla, CA, 92093
| | - Gary Hardiman
- Department of Medicine, University of California San Diego School of Medicine, La Jolla, CA, 92093
| | - Tannishtha Reya
- Department of Pharmacology, University of California San Diego School of Medicine, La Jolla, CA, 92093
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710
- Moores Cancer Center, University of California San Diego School of Medicine, La Jolla, CA, 92093
| |
Collapse
|
111
|
Reelin in the Years: Controlling Neuronal Migration and Maturation in the Mammalian Brain. ACTA ACUST UNITED AC 2014. [DOI: 10.1155/2014/597395] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The extracellular protein Reelin was initially identified as an essential factor in the control of neuronal migration and layer formation in the developing mammalian brain. In the years following its discovery, however, it became clear that Reelin is a multifunctional protein that controls not only the positioning of neurons in the developing brain, but also their growth, maturation, and synaptic activity in the adult brain. In this review, we will highlight the major discoveries of the biological activities of Reelin and the underlying molecular mechanisms that affect the development and function of the mammalian brain, from embryonic ages to adulthood.
Collapse
|
112
|
Klejnot M, Falnikar A, Ulaganathan V, Cross RA, Baas PW, Kozielski F. The crystal structure and biochemical characterization of Kif15: a bifunctional molecular motor involved in bipolar spindle formation and neuronal development. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2014; 70:123-33. [PMID: 24419385 PMCID: PMC3919264 DOI: 10.1107/s1399004713028721] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 10/18/2013] [Indexed: 12/04/2022]
Abstract
Kinesins constitute a superfamily of microtubule-based motor proteins with important cellular functions ranging from intracellular transport to cell division. Some kinesin family members function during the mitotic phase of the eukaryotic cell cycle and are crucial for the successful progression of cell division. In the early stages of mitosis, during prometaphase, certain kinesins are required for the formation of the bipolar spindle, such as Eg5 and Kif15, which seem to possess partially overlapping functions. Because kinesins transform the chemical energy from ATP hydrolysis into mechanical work, inhibition of their function is a tractable approach for drug development. Drugs targeting Eg5 have shown promise as anticancer agents. Kif15 has recently come to the fore because it can substitute the functions of Eg5, and may itself have potential as a prospective drug target. Here, the initial biochemical, kinetic and structural characterization of Kif15 is reported and it is compared with the functionally related motor Eg5. Although Kif15 contains ADP in the catalytic site, its motor-domain structure was captured in the `ATP-like' configuration, with the neck linker docked to the catalytic core. The interaction of Kif15 with microtubules was also investigated and structural differences between these two motors were elucidated which indicate profound differences in their mode of action, in agreement with current models of microtubule cross-linking and sliding.
Collapse
Affiliation(s)
- Marta Klejnot
- The Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, Scotland
| | - Aditi Falnikar
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | | | - Robert A. Cross
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, England
| | - Peter W. Baas
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Frank Kozielski
- School of Pharmacy, University College London, 29–39 Brunswick Square, London WC1N 1AX, England
| |
Collapse
|
113
|
Ramahi JS, Solecki DJ. The PAR polarity complex and cerebellar granule neuron migration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 800:113-31. [PMID: 24243103 DOI: 10.1007/978-94-007-7687-6_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Proper migration of neurons is one of the most important aspects of early brain development. After neuronal progenitors are born in their respective germinal niches, they must migrate to their final locations to form precise neural circuits. A majority of migrating neurons move by associating and disassociating with glial fibers, which serve as scaffolding for the developing brain. Cerebellar granule neurons provide a model system for examination of the mechanisms of neuronal migration in dissociated and slice culture systems; the ability to purify these cells allows migration assays to be paired with genetic, molecular, and biochemical findings. CGNs migrate in a highly polarized fashion along radial glial fibers, using a two-stroke nucleokinesis cycle. The PAR polarity complex of PARD3, PARD6, and an atypical protein kinase C (aPKC) regulate several aspects of neuronal migration. The PAR polarity complex regulates the coordinated movements of the centrosome and soma during nucleokinesis, and also the stability of the microtubule cytoskeleton during migration. PAR proteins coordinate actomyosin dynamics in the leading process of migrating neurons, which are required for migration. The PAR complex also controls the cell-cell adhesions made by migrating neurons along glial cells, and through this mechanism regulates germinal zone exit during prenatal brain development. These findings suggest that the PAR complex coordinates the movement of multiple cellular elements as neurons migrate and that further examination of PAR complex effectors will not only provide novel insights to address fundamental challenges to the field but also expand our understanding of how the PAR complex functions at the molecular level.
Collapse
Affiliation(s)
- Joseph S Ramahi
- Department of Developmental Neurobiology, Saint Jude Children's Research Hospital, MS 325, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | | |
Collapse
|
114
|
Hippenmeyer S. Molecular pathways controlling the sequential steps of cortical projection neuron migration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 800:1-24. [PMID: 24243097 DOI: 10.1007/978-94-007-7687-6_1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Coordinated migration of newly-born neurons to their target territories is essential for correct neuronal circuit assembly in the developing brain. Although a cohort of signaling pathways has been implicated in the regulation of cortical projection neuron migration, the precise molecular mechanisms and how a balanced interplay of cell-autonomous and non-autonomous functions of candidate signaling molecules controls the discrete steps in the migration process, are just being revealed. In this chapter, I will focally review recent advances that improved our understanding of the cell-autonomous and possible cell-nonautonomous functions of the evolutionarily conserved LIS1/NDEL1-complex in regulating the sequential steps of cortical projection neuron migration. I will then elaborate on the emerging concept that the Reelin signaling pathway, acts exactly at precise stages in the course of cortical projection neuron migration. Lastly, I will discuss how finely tuned transcriptional programs and downstream effectors govern particular aspects in driving radial migration at discrete stages and how they regulate the precise positioning of cortical projection neurons in the developing cerebral cortex.
Collapse
Affiliation(s)
- Simon Hippenmeyer
- Developmental Neurobiology, IST Austria (Institute of Science and Technology Austria), Am Campus 1, A-3400, Klosterneuburg, Austria,
| |
Collapse
|
115
|
Li G, Pleasure SJ. The development of hippocampal cellular assemblies. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2013; 3:165-77. [PMID: 24719288 DOI: 10.1002/wdev.127] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Revised: 08/19/2013] [Accepted: 08/29/2013] [Indexed: 11/10/2022]
Abstract
The proper assembly of a cohort of distinct cell types is a prerequisite for building a functional hippocampus. In this review, we describe the major molecular events of the developmental program leading to the cellular construction of the hippocampus. Data from rodent studies are used here to elaborate on our understanding of these processes.
Collapse
Affiliation(s)
- Guangnan Li
- Department of Neurology, Programs in Neuroscience and Developmental Biology, Institute for Regenerative Medicine, University of California, San Francisco, CA, USA
| | | |
Collapse
|
116
|
Germain J, Bruel-Jungerman E, Grannec G, Denis C, Lepousez G, Giros B, Francis F, Nosten-Bertrand M. Doublecortin knockout mice show normal hippocampal-dependent memory despite CA3 lamination defects. PLoS One 2013; 8:e74992. [PMID: 24073232 PMCID: PMC3779246 DOI: 10.1371/journal.pone.0074992] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 08/12/2013] [Indexed: 11/23/2022] Open
Abstract
Mutations in the human X-linked doublecortin gene (DCX) cause major neocortical disorganization associated with severe intellectual disability and intractable epilepsy. Although Dcx knockout (KO) mice exhibit normal isocortical development and architecture, they show lamination defects of the hippocampal pyramidal cell layer largely restricted to the CA3 region. Dcx-KO mice also exhibit interneuron abnormalities. As well as the interest of testing their general neurocognitive profile, Dcx-KO mice also provide a relatively unique model to assess the effects of a disorganized CA3 region on learning and memory. Based on its prominent anatomical and physiological features, the CA3 region is believed to contribute to rapid encoding of novel information, formation and storage of arbitrary associations, novelty detection, and short-term memory. We report here that Dcx-KO adult males exhibit remarkably preserved hippocampal- and CA3-dependant cognitive processes using a large battery of classical hippocampus related tests such as the Barnes maze, contextual fear conditioning, paired associate learning and object recognition. In addition, we show that hippocampal adult neurogenesis, in terms of proliferation, survival and differentiation of granule cells, is also remarkably preserved in Dcx-KO mice. In contrast, following social deprivation, Dcx-KO mice exhibit impaired social interaction and reduced aggressive behaviors. In addition, Dcx-KO mice show reduced behavioral lateralization. The Dcx-KO model thus reinforces the association of neuropsychiatric behavioral impairments with mouse models of intellectual disability.
Collapse
Affiliation(s)
- Johanne Germain
- INSERM UMRS 952, Paris, France
- CNRS UMR 7224, Paris, France
- UPMC, Paris, France
- Université Paris Descartes, Paris, France
| | - Elodie Bruel-Jungerman
- UPMC, Paris, France
- INSERM UMR-S 839, Paris, France
- Institut du Fer à Moulin, Paris, France
| | - Gael Grannec
- INSERM UMRS 952, Paris, France
- CNRS UMR 7224, Paris, France
- UPMC, Paris, France
| | - Cécile Denis
- INSERM UMRS 952, Paris, France
- CNRS UMR 7224, Paris, France
- UPMC, Paris, France
| | | | - Bruno Giros
- INSERM UMRS 952, Paris, France
- CNRS UMR 7224, Paris, France
- UPMC, Paris, France
- Douglas Hospital Research Center, Department of Psychiatry, McGill University, Montreal, Canada
| | - Fiona Francis
- UPMC, Paris, France
- INSERM UMR-S 839, Paris, France
- Institut du Fer à Moulin, Paris, France
| | | |
Collapse
|
117
|
Moon HM, Youn YH, Pemble H, Yingling J, Wittmann T, Wynshaw-Boris A. LIS1 controls mitosis and mitotic spindle organization via the LIS1-NDEL1-dynein complex. Hum Mol Genet 2013; 23:449-66. [PMID: 24030547 DOI: 10.1093/hmg/ddt436] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Heterozygous LIS1 mutations are responsible for the human neuronal migration disorder lissencephaly. Mitotic functions of LIS1 have been suggested from many organisms throughout evolution. However, the cellular functions of LIS1 at distinct intracellular compartments such as the centrosome and the cell cortex have not been well defined especially during mitotic cell division. Here, we used detailed cellular approaches and time-lapse live cell imaging of mitosis from Lis1 mutant mouse embryonic fibroblasts to reveal critical roles of LIS1 in mitotic spindle regulation. We found that LIS1 is required for the tight control of chromosome congression and segregation to dictate kinetochore-microtubule (MT) interactions and anaphase progression. In addition, LIS1 is essential for the establishment of mitotic spindle pole integrity by maintaining normal centrosome number. Moreover, LIS1 plays crucial roles in mitotic spindle orientation by increasing the density of astral MT plus-end movements toward the cell cortex, which enhances cortical targeting of LIS1-dynein complex. Overexpression of NDEL1-dynein and MT stabilization rescues spindle orientation defects in Lis1 mutants, demonstrating that mouse LIS1 acts via the LIS1-NDEL1-dynein complex to regulate astral MT plus-ends dynamics and establish proper contacts of MTs with the cell cortex to ensure precise cell division.
Collapse
Affiliation(s)
- Hyang Mi Moon
- Department of Pediatrics, Institute for Human Genetics
| | | | | | | | | | | |
Collapse
|
118
|
Guijarro P, Wang Y, Ying Y, Yao Y, Jieyi X, Yuan X. In vivoknockdown of ckit impairs neuronal migration and axonal extension in the cerebral cortex. Dev Neurobiol 2013; 73:871-87. [DOI: 10.1002/dneu.22107] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 06/05/2013] [Accepted: 07/02/2013] [Indexed: 12/21/2022]
Affiliation(s)
- Patricia Guijarro
- State Key Laboratory of Neuroscience, Institute of Neuroscience (ION); Shanghai Institutes for Biological Sciences (SIBS); Shanghai 200031 China
- CAS-MPG Partner Institute for Computational Biology (PICB); Shanghai Institutes for Biological Sciences (SIBS); Shanghai 200031 China
| | - Yi Wang
- State Key Laboratory of Neuroscience, Institute of Neuroscience (ION); Shanghai Institutes for Biological Sciences (SIBS); Shanghai 200031 China
| | - Yanting Ying
- State Key Laboratory of Neuroscience, Institute of Neuroscience (ION); Shanghai Institutes for Biological Sciences (SIBS); Shanghai 200031 China
| | - Yini Yao
- State Key Laboratory of Neuroscience, Institute of Neuroscience (ION); Shanghai Institutes for Biological Sciences (SIBS); Shanghai 200031 China
| | - Xiong Jieyi
- CAS-MPG Partner Institute for Computational Biology (PICB); Shanghai Institutes for Biological Sciences (SIBS); Shanghai 200031 China
| | - Xiaobing Yuan
- State Key Laboratory of Neuroscience, Institute of Neuroscience (ION); Shanghai Institutes for Biological Sciences (SIBS); Shanghai 200031 China
| |
Collapse
|
119
|
Zeng L, Wan Y, Li D, Wu J, Shao M, Chen J, Hui L, Ji H, Zhu X. The m subunit of murine translation initiation factor eIF3 maintains the integrity of the eIF3 complex and is required for embryonic development, homeostasis, and organ size control. J Biol Chem 2013; 288:30087-30093. [PMID: 24003236 DOI: 10.1074/jbc.m113.506147] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Mammalian eIF3 is composed of 13 subunits and is the largest eukaryotic initiation factor. eIF3 plays a key role in protein biosynthesis. However, it is not fully understood how different subunits contribute to the structural integrity and function of the eIF3 complex. Whether eIF3 is essential for embryonic development and homeostasis is also not known. Here, we show that eIF3m null embryos are lethal at the peri-implantation stage. Compound heterozygotes (eIF3m(flox)(/-)) or FABP4-Cre-mediated conditional knock-out mice are lethal at mid-gestation stages. Although the heterozygotes are viable, they show markedly reduced organ size and diminished body weight. Acute ablation of eIF3m in adult mouse liver leads to rapidly decreased body weight and death within 2 weeks; these effects are correlated with a severe decline of protein biogenesis in the liver. Protein analyses reveal that eIF3m deficiency significantly impairs the integrity of the eIF3 complex due to down-regulation of multiple other subunits. Two of the subunits, eIF3f and eIF3h, are stabilized by eIF3m through subcomplex formation. Therefore, eIF3m is required for the structural integrity and translation initiation function of eIF3. Furthermore, not only is eIF3m an essential gene, but its expression level is also important for mouse embryonic development and the control of organ size.
Collapse
Affiliation(s)
- Liyong Zeng
- From the State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, and
| | - Yihan Wan
- From the State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, and
| | - Dan Li
- From the State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, and
| | - Jing Wu
- the Model Animal Research Center, Nanjing University, Nanjing, Jiangsu 210061, China
| | - Mengle Shao
- the Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China and
| | - Jiong Chen
- the Model Animal Research Center, Nanjing University, Nanjing, Jiangsu 210061, China
| | - Lijian Hui
- From the State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, and
| | - Hongbin Ji
- From the State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, and
| | - Xueliang Zhu
- From the State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, and.
| |
Collapse
|
120
|
Khalaf-Nazzal R, Bruel-Jungerman E, Rio JP, Bureau J, Irinopoulou T, Sumia I, Roumegous A, Martin E, Olaso R, Parras C, Cifuentes-Diaz C, Francis F. Organelle and cellular abnormalities associated with hippocampal heterotopia in neonatal doublecortin knockout mice. PLoS One 2013; 8:e72622. [PMID: 24023755 PMCID: PMC3759370 DOI: 10.1371/journal.pone.0072622] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 07/11/2013] [Indexed: 11/18/2022] Open
Abstract
Heterotopic or aberrantly positioned cortical neurons are associated with epilepsy and intellectual disability. Various mouse models exist with forms of heterotopia, but the composition and state of cells developing in heterotopic bands has been little studied. Dcx knockout (KO) mice show hippocampal CA3 pyramidal cell lamination abnormalities, appearing from the age of E17.5, and mice suffer from spontaneous epilepsy. The Dcx KO CA3 region is organized in two distinct pyramidal cell layers, resembling a heterotopic situation, and exhibits hyperexcitability. Here, we characterized the abnormally organized cells in postnatal mouse brains. Electron microscopy confirmed that the Dcx KO CA3 layers at postnatal day (P) 0 are distinct and separated by an intermediate layer devoid of neuronal somata. We found that organization and cytoplasm content of pyramidal neurons in each layer were altered compared to wild type (WT) cells. Less regular nuclei and differences in mitochondria and Golgi apparatuses were identified. Each Dcx KO CA3 layer at P0 contained pyramidal neurons but also other closely apposed cells, displaying different morphologies. Quantitative PCR and immunodetections revealed increased numbers of oligodendrocyte precursor cells (OPCs) and interneurons in close proximity to Dcx KO pyramidal cells. Immunohistochemistry experiments also showed that caspase-3 dependent cell death was increased in the CA1 and CA3 regions of Dcx KO hippocampi at P2. Thus, unsuspected ultrastructural abnormalities and cellular heterogeneity may lead to abnormal neuronal function and survival in this model, which together may contribute to the development of hyperexcitability.
Collapse
Affiliation(s)
- Reham Khalaf-Nazzal
- INSERM UMRS 839, Paris, France
- Université Pierre et Marie Curie, Paris, France
- Institut du Fer à Moulin, Paris, France
| | - Elodie Bruel-Jungerman
- INSERM UMRS 839, Paris, France
- Université Pierre et Marie Curie, Paris, France
- Institut du Fer à Moulin, Paris, France
| | - Jean-Paul Rio
- INSERM UMRS 839, Paris, France
- Université Pierre et Marie Curie, Paris, France
- Institut du Fer à Moulin, Paris, France
| | - Jocelyne Bureau
- INSERM UMRS 839, Paris, France
- Université Pierre et Marie Curie, Paris, France
- Institut du Fer à Moulin, Paris, France
| | - Theano Irinopoulou
- INSERM UMRS 839, Paris, France
- Université Pierre et Marie Curie, Paris, France
- Institut du Fer à Moulin, Paris, France
| | - Iffat Sumia
- INSERM UMRS 839, Paris, France
- Université Pierre et Marie Curie, Paris, France
- Institut du Fer à Moulin, Paris, France
| | - Audrey Roumegous
- INSERM UMRS 839, Paris, France
- Université Pierre et Marie Curie, Paris, France
- Institut du Fer à Moulin, Paris, France
| | - Elodie Martin
- Université Pierre et Marie Curie, Paris, France
- Centre de Recherche de l'Institut du Cerveau et de la Moelle épinière, Paris, France
- INSERM UMRS 975, Paris, France
- CNRS UMR 7225, Paris, France
| | - Robert Olaso
- Plateforme de Transcriptomique, Laboratoire de Recherche Translationnelle, CEA/DSV/IG-Centre National de Génotypage, Evry, France
| | - Carlos Parras
- Université Pierre et Marie Curie, Paris, France
- Centre de Recherche de l'Institut du Cerveau et de la Moelle épinière, Paris, France
- INSERM UMRS 975, Paris, France
- CNRS UMR 7225, Paris, France
| | - Carmen Cifuentes-Diaz
- INSERM UMRS 839, Paris, France
- Université Pierre et Marie Curie, Paris, France
- Institut du Fer à Moulin, Paris, France
- * E-mail: (FF); (CCD)
| | - Fiona Francis
- INSERM UMRS 839, Paris, France
- Université Pierre et Marie Curie, Paris, France
- Institut du Fer à Moulin, Paris, France
- * E-mail: (FF); (CCD)
| |
Collapse
|
121
|
Reiner O, Sapir T. LIS1 functions in normal development and disease. Curr Opin Neurobiol 2013; 23:951-6. [PMID: 23973156 DOI: 10.1016/j.conb.2013.08.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 08/01/2013] [Accepted: 08/01/2013] [Indexed: 10/26/2022]
Abstract
LIS1, the first gene to be identified as involved in a neuronal migration disease, is a dosage-sensitive gene whose proper levels are required for multiple aspects of cortical development. Deletions in LIS1 result in a severe brain malformation, known as lissencephaly, whereas duplications delay brain development. LIS1 affects the proliferation of progenitors, spindle orientation and interkinetic nuclear movement in the ventricular zone, as well as nucleokinesis and migration of neurons. LIS1 regulatory interaction with the minus end directed molecular motor cytoplasmic dynein is the key for understanding its complex cellular functions. LIS1-dynein interaction decreases the average velocity of the molecular motor in vitro, shows more complex effects in vivo, and may be of importance in high-load transport especially in neurons.
Collapse
Affiliation(s)
- Orly Reiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel.
| | | |
Collapse
|
122
|
Andersen TA, Troelsen KDLL, Larsen LA. Of mice and men: molecular genetics of congenital heart disease. Cell Mol Life Sci 2013; 71:1327-52. [PMID: 23934094 PMCID: PMC3958813 DOI: 10.1007/s00018-013-1430-1] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 07/16/2013] [Accepted: 07/18/2013] [Indexed: 12/21/2022]
Abstract
Congenital heart disease (CHD) affects nearly 1 % of the population. It is a complex disease, which may be caused by multiple genetic and environmental factors. Studies in human genetics have led to the identification of more than 50 human genes, involved in isolated CHD or genetic syndromes, where CHD is part of the phenotype. Furthermore, mapping of genomic copy number variants and exome sequencing of CHD patients have led to the identification of a large number of candidate disease genes. Experiments in animal models, particularly in mice, have been used to verify human disease genes and to gain further insight into the molecular pathology behind CHD. The picture emerging from these studies suggest that genetic lesions associated with CHD affect a broad range of cellular signaling components, from ligands and receptors, across down-stream effector molecules to transcription factors and co-factors, including chromatin modifiers.
Collapse
Affiliation(s)
- Troels Askhøj Andersen
- Wilhelm Johannsen Centre for Functional Genome Research, Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | | | | |
Collapse
|
123
|
Dix CI, Soundararajan HC, Dzhindzhev NS, Begum F, Suter B, Ohkura H, Stephens E, Bullock SL. Lissencephaly-1 promotes the recruitment of dynein and dynactin to transported mRNAs. J Cell Biol 2013; 202:479-94. [PMID: 23918939 PMCID: PMC3734092 DOI: 10.1083/jcb.201211052] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 06/19/2013] [Indexed: 11/22/2022] Open
Abstract
Microtubule-based transport mediates the sorting and dispersal of many cellular components and pathogens. However, the mechanisms by which motor complexes are recruited to and regulated on different cargos remain poorly understood. Here we describe a large-scale biochemical screen for novel factors associated with RNA localization signals mediating minus end-directed mRNA transport during Drosophila development. We identified the protein Lissencephaly-1 (Lis1) and found that minus-end travel distances of localizing transcripts are dramatically reduced in lis1 mutant embryos. Surprisingly, given its well-documented role in regulating dynein mechanochemistry, we uncovered an important requirement for Lis1 in promoting the recruitment of dynein and its accessory complex dynactin to RNA localization complexes. Furthermore, we provide evidence that Lis1 levels regulate the overall association of dynein with dynactin. Our data therefore reveal a critical role for Lis1 within the mRNA localization machinery and suggest a model in which Lis1 facilitates motor complex association with cargos by promoting the interaction of dynein with dynactin.
Collapse
Affiliation(s)
- Carly I. Dix
- Cell Biology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, England, UK
| | | | - Nikola S. Dzhindzhev
- The Wellcome Trust Centre for Cell Biology, The University of Edinburgh, Edinburgh EH9 3JR, Scotland, UK
| | - Farida Begum
- Cell Biology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, England, UK
| | - Beat Suter
- Institute of Cell Biology, University of Bern, 3012 Bern, Switzerland
| | - Hiroyuki Ohkura
- The Wellcome Trust Centre for Cell Biology, The University of Edinburgh, Edinburgh EH9 3JR, Scotland, UK
| | - Elaine Stephens
- Cell Biology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, England, UK
| | - Simon L. Bullock
- Cell Biology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, England, UK
| |
Collapse
|
124
|
Lanctot AA, Peng CY, Pawlisz AS, Joksimovic M, Feng Y. Spatially dependent dynamic MAPK modulation by the Nde1-Lis1-Brap complex patterns mammalian CNS. Dev Cell 2013; 25:241-55. [PMID: 23673330 DOI: 10.1016/j.devcel.2013.04.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 01/18/2013] [Accepted: 04/09/2013] [Indexed: 02/07/2023]
Abstract
Regulating cell proliferation and differentiation in CNS development requires both extraordinary complexity and precision. Neural progenitors receive graded overlapping signals from midline signaling centers, yet each makes a unique cell fate decision in a spatiotemporally restricted pattern. The Nde1-Lis1 complex regulates individualized cell fate decisions based on the geographical location with respect to the midline. While cells distant from the midline fail to self-renew in the Nde1-Lis1 double-mutant CNS, cells embedded in the signaling centers showed marked overproliferation. A direct interaction between Lis1 and Brap, a mitogen-activated protein kinase (MAPK) signaling threshold modulator, mediates this differential response to mitogenic signal gradients. Nde1-Lis1 deficiency resulted in a spatially dependent alteration of MAPK scaffold Ksr and hyperactivation of MAPK. Epistasis analyses supported synergistic Brap and Lis1 functions. These results suggest that a molecular complex composed of Nde1, Lis1, and Brap regulates the dynamic MAPK signaling threshold in a spatially dependent fashion.
Collapse
Affiliation(s)
- Alison A Lanctot
- Department of Neurology, Northwestern University Feinberg School of Medicine, 303 E. Superior Street, Chicago, IL 60611, USA
| | | | | | | | | |
Collapse
|
125
|
Shyamasundar S, Jadhav SP, Bay BH, Tay SSW, Kumar SD, Rangasamy D, Dheen ST. Analysis of epigenetic factors in mouse embryonic neural stem cells exposed to hyperglycemia. PLoS One 2013; 8:e65945. [PMID: 23776576 PMCID: PMC3679101 DOI: 10.1371/journal.pone.0065945] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 04/30/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Maternal diabetes alters gene expression leading to neural tube defects (NTDs) in the developing brain. The mechanistic pathways that deregulate the gene expression remain unknown. It is hypothesized that exposure of neural stem cells (NSCs) to high glucose/hyperglycemia results in activation of epigenetic mechanisms which alter gene expression and cell fate during brain development. METHODS AND FINDINGS NSCs were isolated from normal pregnancy and streptozotocin induced-diabetic pregnancy and cultured in physiological glucose. In order to examine hyperglycemia induced epigenetic changes in NSCs, chromatin reorganization, global histone status at lysine 9 residue of histone H3 (acetylation and trimethylation) and global DNA methylation were examined and found to be altered by hyperglycemia. In NSCs, hyperglycemia increased the expression of Dcx (Doublecortin) and Pafah1b1 (Platelet activating factor acetyl hydrolase, isoform 1b, subunit 1) proteins concomitant with decreased expression of four microRNAs (mmu-miR-200a, mmu-miR-200b, mmu-miR-466a-3p and mmu-miR-466 d-3p) predicted to target these genes. Knockdown of specific microRNAs in NSCs resulted in increased expression of Dcx and Pafah1b1 proteins confirming target prediction and altered NSC fate by increasing the expression of neuronal and glial lineage markers. CONCLUSION/INTERPRETATION This study revealed that hyperglycemia alters the epigenetic mechanisms in NSCs, resulting in altered expression of some development control genes which may form the basis for the NTDs. Since epigenetic changes are reversible, they may be valuable therapeutic targets in order to improve fetal outcomes in diabetic pregnancy.
Collapse
Affiliation(s)
- Sukanya Shyamasundar
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | | | | | | | | | | |
Collapse
|
126
|
Sipe CW, Liu L, Lee J, Grimsley-Myers C, Lu X. Lis1 mediates planar polarity of auditory hair cells through regulation of microtubule organization. Development 2013; 140:1785-95. [PMID: 23533177 DOI: 10.1242/dev.089763] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The V-shaped hair bundles atop auditory hair cells and their uniform orientation are manifestations of epithelial planar cell polarity (PCP) required for proper perception of sound. PCP is regulated at the tissue level by a conserved core Wnt/PCP pathway. However, the hair cell-intrinsic polarity machinery is poorly understood. Recent findings implicate hair cell microtubules in planar polarization of hair cells. To elucidate the microtubule-mediated polarity pathway, we analyzed Lis1 function in the auditory sensory epithelium in the mouse. We show that conditional deletion of Lis1 in developing hair cells causes defects in cytoplasmic dynein and microtubule organization, resulting in planar polarity defects without overt effects on the core PCP pathway. Lis1 ablation during embryonic development results in defects in hair bundle morphology and orientation, cellular organization and junctional nectin localization. We present evidence that Lis1 regulates localized Rac-PAK signaling in embryonic hair cells, probably through microtubule-associated Tiam1, a guanine nucleotide exchange factor for Rac. Lis1 ablation in postnatal hair cells significantly disrupts centrosome anchoring and the normal V-shape of hair bundles, accompanied by defects in the pericentriolar matrix and microtubule organization. Lis1 is also required for proper positioning of the Golgi complex and mitochondria as well as for hair cell survival. Together, our results demonstrate that Lis1 mediates the planar polarity of hair cells through regulation of microtubule organization downstream of the tissue polarity pathway.
Collapse
Affiliation(s)
- Conor W Sipe
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | | | | | | | | |
Collapse
|
127
|
Escamez T, Bahamonde O, Tabares-Seisdedos R, Vieta E, Martinez S, Echevarria D. Developmental dynamics of PAFAH1B subunits during mouse brain development. J Comp Neurol 2013; 520:3877-94. [PMID: 22522921 DOI: 10.1002/cne.23128] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Platelet-activating factor (PAF) mediates an array of biological processes in the mammalian central nervous system as a bioactive lipid messenger in synaptic function and dysfunction (plasticity, memory, and neurodegeneration). The intracellular enzyme that deacetylates the PAF (PAFAH1B) is composed of a tetramer of two catalytic subunits, ALPHA1 (PAFAH1B3) and ALPHA2 (PAFAH1B2), and a regulatory dimer of LIS1 (PAFAH1B1). We have investigated the mouse PAFAH1B subunit genes during brain development in normal mice and in mice with a hypomorphic allele for Lis1 (Lis1/sLis1; Cahana et al. [2001] Proc Natl Acad Sci U S A 98:6429-6434). We have analyzed quantitatively (by means of real-time polymerase chain reaction) and qualitatively (by in situ hybridization techniques) the amounts and expression patterns of their transcription in developing and postnatal brain, focusing mainly on differences in two laminated encephalic regions, the forebrain (telencephalon) and hindbrain (cerebellum) separately. The results revealed significant differences in cDNA content between these two brain subdivisions but, more importantly, between the LIS1 complex subunits. In addition, we found significant spatial differences in gene expression patterns. Comparison of results obtained with Lis1/sLis1 analysis also revealed significant temporal and spatial differences in Alpha1 and Lis1 expression levels. Thus, small changes in the amount of the Lis1 gene may differentially regulate expression of Alpha1 and Alpha2, depending on the brain region, which suggests different roles for each LIS1 complex subunit during neural differentiation and neural migration.
Collapse
Affiliation(s)
- Teresa Escamez
- Unidad Mixta de Investigación UVEG-UMH-CIBERSAM, Centro de Investigación Biomédica en Red en el Area de Salud Mental, 03550 San Juan de Alicante, Spain
| | | | | | | | | | | |
Collapse
|
128
|
Wang Y, Lin L, Lai H, Parada LF, Lei L. Transcription factor Sox11 is essential for both embryonic and adult neurogenesis. Dev Dyn 2013; 242:638-53. [PMID: 23483698 DOI: 10.1002/dvdy.23962] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 03/02/2013] [Accepted: 03/02/2013] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Neurogenesis requires neural progenitor cell (NPC) proliferation, neuronal migration, and differentiation. During embryonic development, neurons are generated in specific areas of the developing neuroepithelium and migrate to their appropriate positions. In the adult brain, neurogenesis continues in the subgranular zone (SGZ) of the hippocampal dentate gyrus and the subventricular zone (SVZ) of the lateral ventricle. Although neurogenesis is fundamental to brain development and function, our understanding of the molecular mechanisms that regulate neurogenesis is still limited. RESULTS In this study, we generated a Sox11 floxed allele and a Sox11 null allele in mice using the Cre-loxP technology. We first analyzed the role of the transcription factor Sox11 in embryonic neurogenesis using Sox11 null embryos. We also examined the role of Sox11 in adult hippocampal neurogenesis using Sox11 conditional knockout mice in which Sox11 is specifically deleted in adult NPCs. Sox11 null embryos developed small and disorganized brains, accompanied by transient proliferation deficits in NPCs. Deletion of Sox11 in adult NPCs blunted proliferation in the SGZ. Using functional genomics, we identified potential downstream target genes of Sox11. CONCLUSIONS Taken together, our work provides evidence that Sox11 is required for both embryonic and adult neurogenesis, and identifies potential downstream target genes.
Collapse
Affiliation(s)
- Yong Wang
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
| | | | | | | | | |
Collapse
|
129
|
Shao CY, Zhu J, Xie YJ, Wang Z, Wang YN, Wang Y, Su LD, Zhou L, Zhou TH, Shen Y. Distinct functions of nuclear distribution proteins LIS1, Ndel1 and NudCL in regulating axonal mitochondrial transport. Traffic 2013; 14:785-97. [PMID: 23551859 DOI: 10.1111/tra.12070] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Revised: 03/26/2013] [Accepted: 03/30/2013] [Indexed: 11/27/2022]
Abstract
Neurons critically depend on the long-distance transport of mitochondria. Motor proteins kinesin and dynein control anterograde and retrograde mitochondrial transport, respectively in axons. The regulatory molecules that link them to mitochondria need to be better characterized. Nuclear distribution (Nud) family proteins LIS1, Ndel1 and NudCL are critical components of cytoplasmic dynein complex. Roles of these Nud proteins in neuronal mitochondrial transport are unknown. Here we report distinct functions of LIS1, Ndel1 and NudCL on axonal mitochondrial transport in cultured hippocampal neurons. We found that LIS1 interacted with kinsein family protein KIF5b. Depletion of LIS1 enormously suppressed mitochondrial motility in both anterograde and retrograde directions. Inhibition of either Ndel1 or NudCL only partially reduced retrograde mitochondrial motility. However, knocking down both Ndel1 and NudCL almost blocked retrograde mitochondrial transport, suggesting these proteins may work together to regulate retrograde mitochondrial transport through linking dynein-LIS1 complex. Taken together, our results uncover novel roles of LIS1, Ndel1 and NudCL in the transport of mitochondria in axons.
Collapse
Affiliation(s)
- Chong-Yu Shao
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P. R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Reiner O. LIS1 and DCX: Implications for Brain Development and Human Disease in Relation to Microtubules. SCIENTIFICA 2013; 2013:393975. [PMID: 24278775 PMCID: PMC3820303 DOI: 10.1155/2013/393975] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 02/07/2013] [Indexed: 05/29/2023]
Abstract
Proper lamination of the cerebral cortex requires the orchestrated motility of neurons from their place of birth to their final destination. Improper neuronal migration may result in a wide range of diseases, including brain malformations, such as lissencephaly, mental retardation, schizophrenia, and autism. Ours and other studies have implicated that microtubules and microtubule-associated proteins play an important role in the regulation of neuronal polarization and neuronal migration. Here, we will review normal processes of brain development and neuronal migration, describe neuronal migration diseases, and will focus on the microtubule-associated functions of LIS1 and DCX, which participate in the regulation of neuronal migration and are involved in the human developmental brain disease, lissencephaly.
Collapse
Affiliation(s)
- Orly Reiner
- Department of Molecular Genetics, The Weizmann Institute of Science, 76100 Rehovot, Israel
| |
Collapse
|
131
|
Sudarov A, Gooden F, Tseng D, Gan WB, Ross ME. Lis1 controls dynamics of neuronal filopodia and spines to impact synaptogenesis and social behaviour. EMBO Mol Med 2013; 5:591-607. [PMID: 23483716 PMCID: PMC3628102 DOI: 10.1002/emmm.201202106] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 01/29/2013] [Accepted: 01/31/2013] [Indexed: 11/08/2022] Open
Abstract
LIS1 (PAFAH1B1) mutation can impair neuronal migration, causing lissencephaly in humans. LIS1 loss is associated with dynein protein motor dysfunction, and disrupts the actin cytoskeleton through disregulated RhoGTPases. Recently, LIS1 was implicated as an important protein-network interaction node with high-risk autism spectrum disorder genes expressed in the synapse. How LIS1 might participate in this disorder has not been investigated. We examined the role of LIS1 in synaptogenesis of post-migrational neurons and social behaviour in mice. Two-photon imaging of actin-rich dendritic filopodia and spines in vivo showed significant reductions in elimination and turnover rates of dendritic protrusions of layer V pyramidal neurons in adolescent Lis1+/− mice. Lis1+/− filopodia on immature hippocampal neurons in vitro exhibited reduced density, length and RhoA dependent impaired dynamics compared to Lis1+/+. Moreover, Lis1+/− adolescent mice exhibited deficits in social interaction. Lis1 inactivation restricted to the postnatal hippocampus resulted in similar deficits in dendritic protrusion density and social interactions. Thus, LIS1 plays prominently in dendritic filopodia dynamics and spine turnover implicating reduced dendritic spine plasticity as contributing to developmental autistic-like behaviour.
Collapse
Affiliation(s)
- Anamaria Sudarov
- Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, USA
| | | | | | | | | |
Collapse
|
132
|
Kawauchi T, Shikanai M, Kosodo Y. Extra-cell cycle regulatory functions of cyclin-dependent kinases (CDK) and CDK inhibitor proteins contribute to brain development and neurological disorders. Genes Cells 2013; 18:176-94. [PMID: 23294285 PMCID: PMC3594971 DOI: 10.1111/gtc.12029] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 11/26/2012] [Indexed: 12/21/2022]
Abstract
In developing brains, neural progenitors exhibit cell cycle-dependent nuclear movement within the ventricular zone [interkinetic nuclear migration (INM)] and actively proliferate to produce daughter progenitors and/or neurons, whereas newly generated neurons exit from the cell cycle and begin pial surface-directed migration and maturation. Dysregulation of the balance between the proliferation and the cell cycle exit in neural progenitors is one of the major causes of microcephaly (small brain). Recent studies indicate that cell cycle machinery influences not only the proliferation but also INM in neural progenitors. Furthermore, several cell cycle-related proteins, including p27(kip1) , p57(kip2) , Cdk5, and Rb, regulate the migration of neurons in the postmitotic state, suggesting that the growth arrest confers dual functions on cell cycle regulators. Consistently, several types of microcephaly occur in conjunction with neuronal migration disorders, such as periventricular heterotopia and lissencephaly. However, cell cycle re-entry by disturbance of growth arrest in mature neurons is thought to trigger neuronal cell death in Alzheimer's disease. In this review, we introduce the cell cycle protein-mediated regulation of two types of nuclear movement, INM and neuronal migration, during cerebral cortical development, and discuss the roles of growth arrest in cortical development and neurological disorders.
Collapse
Affiliation(s)
- Takeshi Kawauchi
- Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), Saitama, 332-0012, Japan.
| | | | | |
Collapse
|
133
|
Vagus nerve stimulation inhibits seizure activity and protects blood–brain barrier integrity in kindled rats with cortical dysplasia. Life Sci 2013; 92:289-97. [DOI: 10.1016/j.lfs.2013.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2012] [Revised: 12/17/2012] [Accepted: 01/02/2013] [Indexed: 11/21/2022]
|
134
|
Toba S, Tamura Y, Kumamoto K, Yamada M, Takao K, Hattori S, Miyakawa T, Kataoka Y, Azuma M, Hayasaka K, Amamoto M, Tominaga K, Wynshaw-Boris A, Wanibuchi H, Oka Y, Sato M, Kato M, Hirotsune S. Post-natal treatment by a blood-brain-barrier permeable calpain inhibitor, SNJ1945 rescued defective function in lissencephaly. Sci Rep 2013; 3:1224. [PMID: 23390575 PMCID: PMC3565454 DOI: 10.1038/srep01224] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 01/15/2013] [Indexed: 01/26/2023] Open
Abstract
Toward a therapeutic intervention of lissencephaly, we applied a novel calpain inhibitor, SNJ1945. Peri-natal or post-natal treatment with SNJ1945 rescued defective neuronal migration in Lis1+/− mice, impaired behavioral performance and improvement of 18F-FDG uptake. Furthermore, SNJ1945 improved the neural circuit formation and retrograde transport of NFG in Lis1+/− mice. Thus, SNJ1945 is a potential drug for the treatment of human lissencephaly patients.
Collapse
Affiliation(s)
- Shiori Toba
- Department of Genetic Disease Research, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3 Abeno, Osaka 545-8586, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
135
|
LIS1 deficiency promotes dysfunctional synaptic integration of granule cells generated in the developing and adult dentate gyrus. J Neurosci 2012; 32:12862-75. [PMID: 22973010 DOI: 10.1523/jneurosci.1286-12.2012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Type I lissencephaly, a neuronal migration disorder characterized by cognitive disability and refractory epilepsy, is often caused by heterozygous mutations in the LIS1 gene. Histopathologies of malformation-associated epilepsies have been well described, but it remains unclear whether hyperexcitability is attributable to disruptions in neuronal organization or abnormal circuit function. Here, we examined the effect of LIS1 deficiency on excitatory synaptic function in the dentate gyrus of hippocampus, a region believed to serve critical roles in seizure generation and learning and memory. Mice with heterozygous deletion of LIS1 exhibited robust granule cell layer dispersion, and adult-born granule cells labeled with enhanced green fluorescent protein were abnormally positioned in the molecular layer, hilus, and granule cell layer. In whole-cell patch-clamp recordings, reduced LIS1 function was associated with greater excitatory synaptic input to mature granule cells that was consistent with enhanced release probability at glutamatergic synapses. Adult-born granule cells that were ectopically positioned in the molecular layer displayed a more rapid functional maturation and integration into the synaptic network compared with newborn granule cells located in the hilus or granule cell layer or in wild-type controls. In a conditional knock-out mouse, induced LIS1 deficiency in adulthood also enhanced the excitatory input to granule cells in the absence of neuronal disorganization. These findings indicate that disruption of LIS1 has direct effects on excitatory synaptic transmission independent of laminar disorganization, and the ectopic position of adult-born granule cells within a malformed dentate gyrus critically influences their functional maturation and integration.
Collapse
|
136
|
Sebe JY, Bershteyn M, Hirotsune S, Wynshaw-Boris A, Baraban SC. ALLN rescues an in vitro excitatory synaptic transmission deficit in Lis1 mutant mice. J Neurophysiol 2012; 109:429-36. [PMID: 23100132 DOI: 10.1152/jn.00431.2012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
LIS1 gene mutations lead to a rare neurological disorder, classical lissencephaly, characterized by brain malformations, mental retardation, seizures, and premature death. Mice heterozygous for Lis1 (Lis1(+/-)) exhibit cortical malformations, defects in neuronal migration, increased glutamate-mediated synaptic transmission, and spontaneous electrographic seizures. Recent work demonstrated that in utero treatment of Lis1(+/-) mutant dams with ALLN, a calpain inhibitor, partially rescues neuronal migration defects in the offspring. Given the challenges of in utero drug administration, we examined the therapeutic potential of ALLN on postnatal lissencephalic cells. Voltage- and current-clamp studies were performed with acute hippocampal slices obtained from Lis1 mutant mice and age-matched littermate control mice. Specifically, we determined whether postnatal ALLN treatment can reverse excitatory synaptic transmission deficits, namely, an increase in spontaneous and miniature excitatory postsynaptic current (EPSC) frequency, on CA1 pyramidal neurons observed in tissue slices from Lis1(+/-) mice. We found that acute application of ALLN restored spontaneous and miniature EPSC frequencies to wild-type levels without affecting inhibitory postsynaptic synaptic current. Furthermore, Western blot analysis of protein expression, including proteins involved in excitatory synaptic transmission, demonstrated that ALLN blocks the cleavage of the calpain substrate αII-spectrin but does not rescue Lis1 protein levels in Lis1(+/-) mutants.
Collapse
Affiliation(s)
- Joy Y Sebe
- Epilepsy Research Laboratory, Department of Neurological Surgery, University of California, San Francisco, California 94143, USA
| | | | | | | | | |
Collapse
|
137
|
Borrell V, Reillo I. Emerging roles of neural stem cells in cerebral cortex development and evolution. Dev Neurobiol 2012; 72:955-71. [PMID: 22684946 DOI: 10.1002/dneu.22013] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Expansion and folding of the cerebral cortex are landmark features of mammalian brain evolution, which are recapitulated during embryonic development. Neural stem cells and their derived germinal cells are coordinated during cerebral cortex development to produce the appropriate amounts and types of neurons. This process is further complicated in gyrencephalic species, where newborn neurons must disperse in the tangential axis to expand the cerebral cortex in surface area. Here, we review advances that have been made over the last decade in understanding the nature and diversity of telencephalic neural stem cells and their roles in cortical development, and we discuss recent progress on how newly identified types of cortical progenitor cell populations may have evolved to drive the expansion and folding of the mammalian cerebral cortex.
Collapse
Affiliation(s)
- Víctor Borrell
- Developmental Neurobiology Unit, Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, 03550 Sant Joan d'Alacant, Spain.
| | | |
Collapse
|
138
|
Miyoshi G, Fishell G. Dynamic FoxG1 expression coordinates the integration of multipolar pyramidal neuron precursors into the cortical plate. Neuron 2012; 74:1045-58. [PMID: 22726835 DOI: 10.1016/j.neuron.2012.04.025] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2012] [Indexed: 01/20/2023]
Abstract
Pyramidal cells of the cerebral cortex are born in the ventricular zone and migrate through the intermediate zone to enter into the cortical plate. In the intermediate zone, these migrating precursors move tangentially and initiate the extension of their axons by transiently adopting a characteristic multipolar morphology. We observe that expression of the forkhead transcription factor FoxG1 is dynamically regulated during this transitional period. By utilizing conditional genetic strategies, we show that the downregulation of FoxG1 at the beginning of the multipolar cell phase induces Unc5D expression, the timing of which ultimately determines the laminar identity of pyramidal neurons. In addition, we demonstrate that the re-expression of FoxG1 is required for cells to transit out of the multipolar cell phase and to enter into the cortical plate. Thus, the dynamic expression of FoxG1 during migration within the intermediate zone is essential for the proper assembly of the cerebral cortex.
Collapse
Affiliation(s)
- Goichi Miyoshi
- NYU Neuroscience Institute, Department of Physiology and Neuroscience, Smilow Research Center, New York University School of Medicine, 522 First Avenue, New York, NY 10016, USA
| | | |
Collapse
|
139
|
Rompolas P, Patel-King RS, King SM. Association of Lis1 with outer arm dynein is modulated in response to alterations in flagellar motility. Mol Biol Cell 2012; 23:3554-65. [PMID: 22855525 PMCID: PMC3442404 DOI: 10.1091/mbc.e12-04-0287] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The cytoplasmic dynein regulatory factor Lis1, which induces a persistent tight binding to microtubules and allows for transport of cargoes under high-load conditions, is also present in motile cilia/flagella. Lis1 levels in cilia/flagella are dynamically modulated in response to imposed alterations in beat parameters. The cytoplasmic dynein regulatory factor Lis1, which induces a persistent tight binding to microtubules and allows for transport of cargoes under high-load conditions, is also present in motile cilia/flagella. We observed that Lis1 levels in flagella of Chlamydomonas strains that exhibit defective motility due to mutation of various axonemal substructures were greatly enhanced compared with wild type; this increase was absolutely dependent on the presence within the flagellum of the outer arm dynein α heavy chain/light chain 5 thioredoxin unit. To assess whether cells might interpret defective motility as a “high-load environment,” we reduced the flagellar beat frequency of wild-type cells through enhanced viscous load and by reductive stress; both treatments resulted in increased levels of flagellar Lis1, which altered the intrinsic beat frequency of the trans flagellum. Differential extraction of Lis1 from wild-type and mutant axonemes suggests that the affinity of outer arm dynein for Lis1 is directly modulated. In cytoplasm, Lis1 localized to two punctate structures, one of which was located near the base of the flagella. These data reveal that the cell actively monitors motility and dynamically modulates flagellar levels of the dynein regulatory factor Lis1 in response to imposed alterations in beat parameters.
Collapse
Affiliation(s)
- Panteleimon Rompolas
- Department of Molecular, Microbial and Structural Biology, University of Connecticut Health Center, Farmington, CT 06030-3305, USA
| | | | | |
Collapse
|
140
|
Abstract
Adenomatous polyposis coli 2 (APC2) is a family member of APC and mainly expressed in the nervous system. We previously reported that APC2 plays a critical role in axonal projection through the regulation of microtubule stability. Here, we show that a lack of Apc2 induces severe laminary defects in some regions of the mouse brain, including the cerebral cortex and cerebellum. In vivo BrdU labeling and immunohistochemical analyses with specific markers revealed that the laminary abnormalities are a result of dysregulated neuronal migration by a cell-autonomous mechanism. Using total internal reflection fluorescent microscopy, we found that APC2 is distributed along actin fibers as well as microtubules. Cerebellar granule cells in dissociated cultures and in vivo showed that BDNF-stimulated directional migration is impaired in Apc2-deficient neurons. We revealed that this impairment stems from the dysregulations of Rho family GTPase activation and TrkB localization, which disrupts the formation of BDNF-stimulated F-actin at the leading edge. Thus, APC2 is an essential mediator of the cytoskeletal regulation at leading edges in response to extracellular signals.
Collapse
|
141
|
Sitaram P, Anderson MA, Jodoin JN, Lee E, Lee LA. Regulation of dynein localization and centrosome positioning by Lis-1 and asunder during Drosophila spermatogenesis. Development 2012; 139:2945-54. [PMID: 22764052 DOI: 10.1242/dev.077511] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Dynein, a microtubule motor complex, plays crucial roles in cell-cycle progression in many systems. The LIS1 accessory protein directly binds dynein, although its precise role in regulating dynein remains unclear. Mutation of human LIS1 causes lissencephaly, a developmental brain disorder. To gain insight into the in vivo functions of LIS1, we characterized a male-sterile allele of the Drosophila homolog of human LIS1. We found that centrosomes do not properly detach from the cell cortex at the onset of meiosis in most Lis-1 spermatocytes; centrosomes that do break cortical associations fail to attach to the nucleus. In Lis-1 spermatids, we observed loss of attachments between the nucleus, basal body and mitochondria. The localization pattern of LIS-1 protein throughout Drosophila spermatogenesis mirrors that of dynein. We show that dynein recruitment to the nuclear surface and spindle poles is severely reduced in Lis-1 male germ cells. We propose that Lis-1 spermatogenesis phenotypes are due to loss of dynein regulation, as we observed similar phenotypes in flies null for Tctex-1, a dynein light chain. We have previously identified asunder (asun) as another regulator of dynein localization and centrosome positioning during Drosophila spermatogenesis. We now report that Lis-1 is a strong dominant enhancer of asun and that localization of LIS-1 in male germ cells is ASUN dependent. We found that Drosophila LIS-1 and ASUN colocalize and coimmunoprecipitate from transfected cells, suggesting that they function within a common complex. We present a model in which Lis-1 and asun cooperate to regulate dynein localization and centrosome positioning during Drosophila spermatogenesis.
Collapse
Affiliation(s)
- Poojitha Sitaram
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, U-4225 Medical Research Building III, 465 21st Avenue South, Nashville, TN 37232-8240, USA
| | | | | | | | | |
Collapse
|
142
|
Lo FY, Chen HT, Cheng HC, Hsu HS, Wang YC. Overexpression of PAFAH1B1 is associated with tumor metastasis and poor survival in non-small cell lung cancer. Lung Cancer 2012; 77:585-92. [PMID: 22749159 DOI: 10.1016/j.lungcan.2012.05.105] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 04/11/2012] [Accepted: 05/25/2012] [Indexed: 11/15/2022]
Abstract
Our previous array-comparative genomic hybridization study showed that PAFAH1B1 gene locus was amplified in lung cancer patients, suggesting that PAFAH1B1 is a potential oncogene in lung cancer. Here, we investigate the oncogenic mechanisms of PAFAH1B1 in lung cancer. PAFAH1B1 was characterized in cell and animal models of lung cancer by in vitro migration and invasion assays and in vivo metastasis studies. The mRNA and protein expression levels of PAFAH1B1 were further determined and the prognostic effects of PAFAH1B1 overexpression in lung cancer patients were analyzed. Overexpression of PAFAH1B1 enhanced migration and invasion in lung cancer cells, whereas knockdown of PAFAH1B1 decreased cell migration and invasion, and disrupted cell microtubule organization and pericellular poly-fibronectin assemblies. In vivo tumor metastasis assay confirmed that PAFAH1B1 knockdown in lung cancer cells markedly reduced their metastasis capabilities in animals. The frequencies of overexpressed PAFAH1B1 mRNA and protein were 62.4% (63/101) and 57.4% (58/101) in lung cancer patients, respectively. The clinical correlation results showed that overexpression of PAFAH1B1 was significantly associated with late stage (mRNA: P=0.008, protein: P=0.008) and poor survival in lung adenocarcinoma (P=0.020) and male patients (P=0.049). Our results provide the first evidence that PAFAH1B1 overexpression contributes to lung tumorigenesis and poor prognosis. These effects are partly mediated through disruption of microtubule network and pericellular poly-fibronectin assembly to promote migration and invasiveness of lung cancer cells.
Collapse
Affiliation(s)
- Fang-Yi Lo
- Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | |
Collapse
|
143
|
Egan MJ, Tan K, Reck-Peterson SL. Lis1 is an initiation factor for dynein-driven organelle transport. ACTA ACUST UNITED AC 2012; 197:971-82. [PMID: 22711696 PMCID: PMC3384415 DOI: 10.1083/jcb.201112101] [Citation(s) in RCA: 146] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The dynein-associated protein Lis1 may be a ubiquitous determinant of dynein-dependent transport required primarily at the stage of motility initiation. The molecular motor cytoplasmic dynein is responsible for most minus-end–directed, microtubule-based transport in eukaryotic cells. It is especially important in neurons, where defects in microtubule-based motility have been linked to neurological diseases. For example, lissencephaly is caused by mutations in the dynein-associated protein Lis1. In this paper, using the long, highly polarized hyphae of the filamentous fungus Aspergillus nidulans, we show that three morphologically and functionally distinct dynein cargos showed transport defects in the genetic absence of Lis1/nudF, raising the possibility that Lis1 is ubiquitously used for dynein-based transport. Surprisingly, both dynein and its cargo moved at normal speeds in the absence of Lis1 but with reduced frequency. Moreover, Lis1, unlike dynein and dynactin, was absent from moving dynein cargos, further suggesting that Lis1 is not required for dynein-based cargo motility once it has commenced. Based on these observations, we propose that Lis1 has a general role in initiating dynein-driven motility.
Collapse
Affiliation(s)
- Martin J Egan
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | | | | |
Collapse
|
144
|
Spindle orientation in mammalian cerebral cortical development. Curr Opin Neurobiol 2012; 22:737-46. [PMID: 22554882 PMCID: PMC3500682 DOI: 10.1016/j.conb.2012.04.003] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 04/07/2012] [Indexed: 12/24/2022]
Abstract
In any mitotic cell, the orientation of the mitotic spindle determines the orientation of the cleavage plane and therefore the position of the two daughter cells. When combined with polarization of cellular components, spindle orientation is also a well-conserved means of generating daughter cells with asymmetric cell fates, such as progenitors and differentiated cell types. In the mammalian neocortex, the precise planar spindle orientation observed early during development is vital for symmetric proliferative divisions. During later stages, spindles can be oblique or even vertical but the role of this reorientation is somewhat less clear as asymmetric cell fates can arise independently of spindle orientation during this stage. Although decades of work have identified many key conserved regulators of spindle positioning, its precise role in cell fate determination in the mammalian neocortex has been enigmatic. Recent work focused on mInsc and LGN has now revealed an important role for spindle orientation in determination of specific asymmetric cell fates, namely intermediate progenitors and a new progenitor population, termed outer radial glia. In this way, spindle orientation helps determine the neurogenic outcome of asymmetric progenitor divisions, thereby influencing neuron output and cerebral cortical expansion.
Collapse
|
145
|
Mullen MP, Elia G, Hilliard M, Parr MH, Diskin MG, Evans ACO, Crowe MA. Proteomic characterization of histotroph during the preimplantation phase of the estrous cycle in cattle. J Proteome Res 2012; 11:3004-18. [PMID: 22463384 DOI: 10.1021/pr300144q] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Uterine secretions, or histotroph, are a critical component for early embryo survival, functioning as the sole supply of vitamins, minerals, enzymes, and other myriad of nutrients required by the developing conceptus before implantation. Histotroph is therefore a promising source for biomarkers of uterine function and for enhancing our understanding of the environment supporting early embryo development and survival. Utilizing label-free liquid chromatography-tandem mass spectrometry (LC-MS/MS) shotgun proteomics, we characterized the uterine proteome at two key preimplantation stages of the estrous cycle in high fertility cattle. We identified 300 proteins on Day 7 and 510 proteins on Day 13 including 281 proteins shared between days. Five proteins were more abundant (P < 0.05) on Day 7 compared with Day 13 and included novel histotroph proteins cytokeratin 10 and stathmin. Twenty-nine proteins were more abundant (P < 0.05) including 13 unique on Day 13 compared with Day 7 and included previously identified legumain, metalloprotease inhibitor-2, and novel histotroph proteins chromogranin A and pyridoxal kinase. Functional analysis of the 34 differentially expressed proteins (including 14 novel to histotroph) revealed distinct biological roles putatively involved in early pregnancy, including remodelling of the uterine environment in preparation for implantation; nutrient metabolism; embryo growth, development and protection; maintenance of uterine health; and maternal immune modulation. This study is the first reported LC-MS/MS based global proteomic characterization of the uterine environment in any domesticated species before implantation and provides novel information on the temporal alterations in histotroph composition during critical stages for early embryo development and uterine function during the early establishment of pregnancy.
Collapse
Affiliation(s)
- Michael P Mullen
- Animal and Bioscience Research Department, Animal & Grassland Research and Innovation Centre, Teagasc, Mellows Campus, Athenry, Co. Galway, Ireland.
| | | | | | | | | | | | | |
Collapse
|
146
|
Moore KD, Chen R, Cilluffo M, Golden JA, Phelps PE. Lis1 reduction causes tangential migratory errors in mouse spinal cord. J Comp Neurol 2012; 520:1198-211. [PMID: 21935943 PMCID: PMC4079006 DOI: 10.1002/cne.22768] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mutations in human LIS1 cause abnormal neuronal migration and a smooth brain phenotype known as lissencephaly. Lis1+/− (Pafah1b1) mice show defective lamination in the cerebral cortex and hippocampal formation, whereas homozygous mutations result in embryonic lethality. Given that Lis1 is highly expressed in embryonic neurons, we hypothesized that sympathetic and parasympathetic preganglionic neurons (SPNs and PPNs) would exhibit migratory defects in Lis1+/− mice. The initial radial migration of SPNs and PPNs that occurs together with somatic motor neurons appeared unaffected in Lis1+/− mice. The subsequent dorsally directed tangential migration, however, was aberrant in a subset of these neurons. At all embryonic ages analyzed, the distribution of SPNs and PPNs in Lis1+/− mice was elongated dorsoventrally compared with Lis1+/+ mice. Individual cell bodies of ectopic preganglionic neurons were found in the ventral spinal cord with their leading processes oriented along their dorsal migratory trajectory. By birth, Lis1+/− SPNs and PPNs were separated into distinct groups, those that were correctly, and those incorrectly positioned in the intermediate horn. As mispositioned SPNs and PPNs still were detected in P30 Lis1+/− mice, we conclude that these neurons ceased migration prematurely. Additionally, we found that a dorsally located group of somatic motor neurons in the lumbar spinal cord, the retrodorsolateral nucleus, showed delayed migration in Lis1+/− mice. These results suggest that Lis1 is required for the dorsally directed tangential migration of many sympathetic and parasympathetic preganglionic neurons and a subset of somatic motor neurons.
Collapse
Affiliation(s)
- Katherine D. Moore
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, California 90095-7239
| | - Renee Chen
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, California 90095-7239
| | - Marianne Cilluffo
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, California 90095-7239
| | - Jeffrey A. Golden
- Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia and the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104
| | - Patricia E. Phelps
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, California 90095-7239
| |
Collapse
|
147
|
Cheah PS, Ramshaw HS, Thomas PQ, Toyo-Oka K, Xu X, Martin S, Coyle P, Guthridge MA, Stomski F, van den Buuse M, Wynshaw-Boris A, Lopez AF, Schwarz QP. Neurodevelopmental and neuropsychiatric behaviour defects arise from 14-3-3ζ deficiency. Mol Psychiatry 2012; 17:451-66. [PMID: 22124272 DOI: 10.1038/mp.2011.158] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Complex neuropsychiatric disorders are believed to arise from multiple synergistic deficiencies within connected biological networks controlling neuronal migration, axonal pathfinding and synapse formation. Here, we show that deletion of 14-3-3ζ causes neurodevelopmental anomalies similar to those seen in neuropsychiatric disorders such as schizophrenia, autism spectrum disorder and bipolar disorder. 14-3-3ζ-deficient mice displayed striking behavioural and cognitive deficiencies including a reduced capacity to learn and remember, hyperactivity and disrupted sensorimotor gating. These deficits are accompanied by subtle developmental abnormalities of the hippocampus that are underpinned by aberrant neuronal migration. Significantly, 14-3-3ζ-deficient mice exhibited abnormal mossy fibre navigation and glutamatergic synapse formation. The molecular basis of these defects involves the schizophrenia risk factor, DISC1, which interacts isoform specifically with 14-3-3ζ. Our data provide the first evidence of a direct role for 14-3-3ζ deficiency in the aetiology of neurodevelopmental disorders and identifies 14-3-3ζ as a central risk factor in the schizophrenia protein interaction network.
Collapse
Affiliation(s)
- P S Cheah
- Department of Human Immunology, Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
|
149
|
Kawabata I, Kashiwagi Y, Obashi K, Ohkura M, Nakai J, Wynshaw-Boris A, Yanagawa Y, Okabe S. LIS1-dependent retrograde translocation of excitatory synapses in developing interneuron dendrites. Nat Commun 2012; 3:722. [PMID: 22395613 PMCID: PMC3316883 DOI: 10.1038/ncomms1736] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 02/07/2012] [Indexed: 11/30/2022] Open
Abstract
Synaptic remodelling coordinated with dendritic growth is essential for proper development of neural connections. After establishment of synaptic contacts, synaptic junctions are thought to become stationary and provide fixed anchoring points for further dendritic growth. However, the possibility of active translocation of synapses along dendritic protrusions, to guide the proper arrangement of synaptic distribution, has not yet been fully investigated. Here we show that immature dendrites of γ-aminobutyric acid-positive interneurons form long protrusions and that these protrusions serve as conduits for retrograde translocation of synaptic contacts to the parental dendrites. This translocation process is dependent on microtubules and the activity of LIS1, an essential regulator of dynein-mediated motility. Suppression of this retrograde translocation results in disorganized synaptic patterns on interneuron dendrites. Taken together, these findings suggest the existence of an active microtubule-dependent mechanism for synaptic translocation that helps in the establishment of proper synaptic distribution on dendrites. Maturation of synaptic junctions is important for proper neuronal connections. Using live cell imaging, Okabe et al. show that interneuron dendrites extend filopodia-like projections and use microtubule-dependent retrograde transport to guide proper synaptic distribution on dendrites.
Collapse
Affiliation(s)
- Izumi Kawabata
- Department of Cellular Neurobiology, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | | | | | | | |
Collapse
|
150
|
A Cdk5-dependent switch regulates Lis1/Ndel1/dynein-driven organelle transport in adult axons. J Neurosci 2012; 31:17207-19. [PMID: 22114287 DOI: 10.1523/jneurosci.4108-11.2011] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Lissencephaly is a human developmental brain abnormality caused by LIS1 haploinsufficiency. This disorder is in large part attributed to altered mitosis and migration in the developing brain. LIS1 and an interacting protein, NDEL1, bind to cytoplasmic dynein, a microtubule motor protein. While the tripartite complex is clearly important for developmental events, we are intrigued by the fact that Lis1 and Ndel1 expression remain high in the adult mouse nervous system. Dynein plays a crucial role in retrograde axonal transport, a process that is used by mature neurons. Here, we monitored acidic organelles moving in axons of adult rat sensory neurons to determine whether Lis1 and Ndel1 contribute to axonal transport. Lis1 RNAi significantly reduced axon transport of these organelles. Ndel1 RNAi had little impact, but combined Lis1 and Ndel1 RNAi caused a more severe phenotype than Lis1 RNAi alone, essentially shutting down transport. Lis1 overexpression stimulated retrograde transport, while a Lis1 dynein-binding mutant severely disrupted transport. Overexpression of Ndel1 or a Lis1 Ndel1-binding mutant only mildly perturbed transport. However, expressing a mutant Ndel1 lacking key phosphorylation sites shut down transport completely, as did a dominant-negative Cdk5 construct. We propose that, in axons, unphosphorylated Ndel1 inhibits the capacity of dynein to transport acidic organelles. Phosphorylation of Ndel1 by Cdk5 not only reduces this inhibition but also allows Lis1 to further stimulate the cargo transport capacity of dynein. Our data raise the possibility that defects in a Lis1/Ndel1 regulatory switch could contribute to neurodegenerative diseases linked to axonal pathology in adults.
Collapse
|