1
|
Puls R, von Haefen C, Bührer C, Endesfelder S. Protective Effect of Dexmedetomidine against Hyperoxia-Damaged Cerebellar Neurodevelopment in the Juvenile Rat. Antioxidants (Basel) 2023; 12:antiox12040980. [PMID: 37107355 PMCID: PMC10136028 DOI: 10.3390/antiox12040980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023] Open
Abstract
Impaired cerebellar development of premature infants and the associated impairment of cerebellar functions in cognitive development could be crucial factors for neurodevelopmental disorders. Anesthetic- and hyperoxia-induced neurotoxicity of the immature brain can lead to learning and behavioral disorders. Dexmedetomidine (DEX), which is associated with neuroprotective properties, is increasingly being studied for off-label use in the NICU. For this purpose, six-day-old Wistar rats (P6) were exposed to hyperoxia (80% O2) or normoxia (21% O2) for 24 h after DEX (5 µg/kg, i.p.) or vehicle (0.9% NaCl) application. An initial detection in the immature rat cerebellum was performed after the termination of hyperoxia at P7 and then after recovery in room air at P9, P11, and P14. Hyperoxia reduced the proportion of Calb1+-Purkinje cells and affected the dendrite length at P7 and/or P9/P11. Proliferating Pax6+-granule progenitors remained reduced after hyperoxia and until P14. The expression of neurotrophins and neuronal transcription factors/markers of proliferation, migration, and survival were also reduced by oxidative stress in different manners. DEX demonstrated protective effects on hyperoxia-injured Purkinje cells, and DEX without hyperoxia modulated neuronal transcription in the short term without any effects at the cellular level. DEX protects hyperoxia-damaged Purkinje cells and appears to differentially affect cerebellar granular cell neurogenesis following oxidative stress.
Collapse
Affiliation(s)
- Robert Puls
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Clarissa von Haefen
- Department of Anesthesiology and Intensive Care Medicine, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Christoph Bührer
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Stefanie Endesfelder
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
2
|
Hawkes R. Cerebellar Patterning Defects in Mutant Mice. Front Neurosci 2021; 15:787425. [PMID: 34955734 PMCID: PMC8692567 DOI: 10.3389/fnins.2021.787425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/01/2021] [Indexed: 11/25/2022] Open
Abstract
The cerebellar cortex is highly compartmentalized and serves as a remarkable model for pattern formation throughout the brain. In brief, the adult cerebellar cortex is subdivided into five anteroposterior units—transverse zones—and subsequently, each zone is divided into ∼20 parasagittal stripes. Zone-and-stripe pattern formation involves the interplay of two parallel developmental pathways—one for inhibitory neurons, the second for excitatory. In the inhibitory pathway, progenitor cells of the 4th ventricle generate the Purkinje cells and inhibitory interneurons. In the excitatory pathway, progenitor cells in the upper rhombic lip give rise to the external granular layer, and subsequently to the granular layer of the adult. Both the excitatory and inhibitory developmental pathways are spatially patterned and the interactions of the two generate the complex topography of the adult. This review briefly describes the cellular and molecular mechanisms that underly zone-and-stripe development with a particular focus on mutations known to interfere with normal cerebellar development and the light they cast on the mechanisms of pattern formation.
Collapse
Affiliation(s)
- Richard Hawkes
- Department of Cell Biology, Cumming School of Medicine, Anatomy and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
3
|
Consalez GG, Goldowitz D, Casoni F, Hawkes R. Origins, Development, and Compartmentation of the Granule Cells of the Cerebellum. Front Neural Circuits 2021; 14:611841. [PMID: 33519389 PMCID: PMC7843939 DOI: 10.3389/fncir.2020.611841] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/17/2020] [Indexed: 12/21/2022] Open
Abstract
Granule cells (GCs) are the most numerous cell type in the cerebellum and indeed, in the brain: at least 99% of all cerebellar neurons are granule cells. In this review article, we first consider the formation of the upper rhombic lip, from which all granule cell precursors arise, and the way by which the upper rhombic lip generates the external granular layer, a secondary germinal epithelium that serves to amplify the upper rhombic lip precursors. Next, we review the mechanisms by which postmitotic granule cells are generated in the external granular layer and migrate radially to settle in the granular layer. In addition, we review the evidence that far from being a homogeneous population, granule cells come in multiple phenotypes with distinct topographical distributions and consider ways in which the heterogeneity of granule cells might arise during development.
Collapse
Affiliation(s)
- G Giacomo Consalez
- Division of Neuroscience, San Raffaele Scientific Institute, San Raffaele University, Milan, Italy
| | - Daniel Goldowitz
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Filippo Casoni
- Division of Neuroscience, San Raffaele Scientific Institute, San Raffaele University, Milan, Italy
| | - Richard Hawkes
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
4
|
Aiello G, Ballabio C, Ruggeri R, Fagnocchi L, Anderle M, Morassut I, Caron D, Garilli F, Gianno F, Giangaspero F, Piazza S, Romanel A, Zippo A, Tiberi L. Truncated BRPF1 Cooperates with Smoothened to Promote Adult Shh Medulloblastoma. Cell Rep 2020; 29:4036-4052.e10. [PMID: 31851932 DOI: 10.1016/j.celrep.2019.11.046] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 05/14/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022] Open
Abstract
The transition of neural progenitors to differentiated postmitotic neurons is mainly considered irreversible in physiological conditions. In the present work, we show that Shh pathway activation through SmoM2 expression promotes postmitotic neurons dedifferentiation, re-entering in the cell cycle and originating medulloblastoma in vivo. Notably, human adult patients present inactivating mutations of the chromatin reader BRPF1 that are associated with SMO mutations and absent in pediatric and adolescent patients. Here, we found that truncated BRPF1 protein, as found in human adult patients, is able to induce medulloblastoma in adult mice upon SmoM2 activation. Indeed, postmitotic neurons re-entered the cell cycle and proliferated as a result of chromatin remodeling of neurons by BRPF1. Our model of brain cancer explains the onset of a subset of human medulloblastoma in adult individuals where granule neuron progenitors are no longer present.
Collapse
Affiliation(s)
- Giuseppe Aiello
- Armenise-Harvard Laboratory of Brain Disorders and Cancer, CIBIO, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Claudio Ballabio
- Armenise-Harvard Laboratory of Brain Disorders and Cancer, CIBIO, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Riccardo Ruggeri
- Armenise-Harvard Laboratory of Brain Disorders and Cancer, CIBIO, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Luca Fagnocchi
- Laboratory of Chromatin Biology & Epigenetics, CIBIO, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Marica Anderle
- Armenise-Harvard Laboratory of Brain Disorders and Cancer, CIBIO, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Ilaria Morassut
- Armenise-Harvard Laboratory of Brain Disorders and Cancer, CIBIO, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Davide Caron
- Armenise-Harvard Laboratory of Brain Disorders and Cancer, CIBIO, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Francesca Garilli
- Armenise-Harvard Laboratory of Brain Disorders and Cancer, CIBIO, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Francesca Gianno
- Department of Radiologic, Oncologic and Anatomo Pathological Sciences, University Sapienza of Rome, Rome, Italy; IRCCS Neuromed, Pozzilli, Isernia, Italy
| | - Felice Giangaspero
- Department of Radiologic, Oncologic and Anatomo Pathological Sciences, University Sapienza of Rome, Rome, Italy; IRCCS Neuromed, Pozzilli, Isernia, Italy
| | - Silvano Piazza
- Bioinformatics Core Facility, CIBIO, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Alessandro Romanel
- Laboratory of Bioinformatics and Computational Genomics, CIBIO, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Alessio Zippo
- Laboratory of Chromatin Biology & Epigenetics, CIBIO, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Luca Tiberi
- Armenise-Harvard Laboratory of Brain Disorders and Cancer, CIBIO, University of Trento, Via Sommarive 9, 38123 Trento, Italy.
| |
Collapse
|
5
|
Lee TY, Cho IS, Bashyal N, Naya FJ, Tsai MJ, Yoon JS, Choi JM, Park CH, Kim SS, Suh-Kim H. ERK Regulates NeuroD1-mediated Neurite Outgrowth via Proteasomal Degradation. Exp Neurobiol 2020; 29:189-206. [PMID: 32606250 PMCID: PMC7344372 DOI: 10.5607/en20021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/16/2020] [Accepted: 06/16/2020] [Indexed: 12/22/2022] Open
Abstract
Neurogenic differentiation 1 (NeuroD1) is a class B basic helix-loop-helix (bHLH) transcription factor and regulates differentiation and survival of neuronal and endocrine cells by means of several protein kinases, including extracellular signal-regulated kinase (ERK). However, the effect of phosphorylation on the functions of NeuroD1 by ERK has sparked controversy based on context-dependent differences across diverse species and cell types. Here, we evidenced that ERK-dependent phosphorylation controlled the stability of NeuroD1 and consequently, regulated proneural activity in neuronal cells. A null mutation at the ERK-dependent phosphorylation site, S274A, increased the half-life of NeuroD1 by blocking its ubiquitin-dependent proteasomal degradation. The S274A mutation did not interfere with either the nuclear translocation of NeuroD1 or its heterodimerization with E47, its ubiquitous partner and class A bHLH transcription factor. However, the S274A mutant increased transactivation of the E-box-mediated gene and neurite outgrowth in F11 neuroblastoma cells, compared to the wild-type NeuroD1. Transcriptome and Gene Ontology enrichment analyses indicated that genes involved in axonogenesis and dendrite development were downregulated in NeuroD1 knockout (KO) mice. Overexpression of the S274A mutant salvaged neurite outgrowth in NeuroD1-deficient mice, whereas neurite outgrowth was minimal with S274D, a phosphomimicking mutant. Our data indicated that a longer protein half-life enhanced the overall activity of NeuroD1 in stimulating downstream genes and neuronal differentiation. We propose that blocking ubiquitin-dependent proteasomal degradation may serve as a strategy to promote neuronal activity by stimulating the expression of neuron-specific genes in differentiating neurons.
Collapse
Affiliation(s)
- Tae-Young Lee
- Department of Anatomy, Ajou University School of Medicine, Suwon 16499, Korea.,Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon 16499, Korea.,Research Center, CelleBrain Ltd., Jeonju 54871, Korea
| | - In-Su Cho
- Department of Anatomy, Ajou University School of Medicine, Suwon 16499, Korea
| | - Narayan Bashyal
- Department of Anatomy, Ajou University School of Medicine, Suwon 16499, Korea.,Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon 16499, Korea
| | - Francisco J Naya
- Department of Biology, Life Science and Engineering Building, Boston University, Boston, MA 00215, USA
| | - Ming-Jer Tsai
- Department of Medicine and Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jeong Seon Yoon
- Department of Anatomy, Ajou University School of Medicine, Suwon 16499, Korea
| | - Jung-Mi Choi
- Department of Anatomy, Ajou University School of Medicine, Suwon 16499, Korea
| | - Chang-Hwan Park
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
| | - Sung-Soo Kim
- Department of Anatomy, Ajou University School of Medicine, Suwon 16499, Korea.,Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon 16499, Korea
| | - Haeyoung Suh-Kim
- Department of Anatomy, Ajou University School of Medicine, Suwon 16499, Korea.,Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon 16499, Korea.,Research Center, CelleBrain Ltd., Jeonju 54871, Korea
| |
Collapse
|
6
|
Hanzel M, Rook V, Wingate RJT. Mitotic granule cell precursors undergo highly dynamic morphological transitions throughout the external germinal layer of the chick cerebellum. Sci Rep 2019; 9:15218. [PMID: 31645601 PMCID: PMC6811643 DOI: 10.1038/s41598-019-51532-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 10/02/2019] [Indexed: 01/16/2023] Open
Abstract
The developing cerebellum of amniotes is characterised by a unique, transient, secondary proliferation zone: the external germinal layer (EGL). The EGL is comprised solely of granule cell precursors, whose progeny migrate inwardly to form the internal granule cell layer. While a range of cell morphologies in the EGL has long been known, how they reflect the cells' differentiation status has previously only been inferred. Observations have suggested a deterministic maturation from outer to inner EGL that we wished to test experimentally. To do this, we electroporated granule cell precursors in chick with plasmids encoding fluorescent proteins and probed labelled cells with markers of both proliferation (phosphohistone H3) and differentiation (Axonin1/TAG1 and NeuroD1). We show that granule cell precursors can display a range of complex forms throughout the EGL while mitotically active. Overexpression of full length NeuroD1 within granule cell precursors does not abolish proliferation, but biases granule cells towards precocious differentiation, alters their migration path and results in a smaller and less foliated cerebellum. Our results show that granule cells show a greater flexibility in differentiation than previously assumed. We speculate that this allows the EGL to regulate its proliferative activity in response to overall patterns of cerebellar growth.
Collapse
Affiliation(s)
- Michalina Hanzel
- MRC Centre for Neurodevelopmental Disorders, King's College London, Institute of Psychiatry, Psychology and Neuroscience, 4th floor New Hunt's House, Guy's Campus, London, UK
| | - Victoria Rook
- MRC Centre for Neurodevelopmental Disorders, King's College London, Institute of Psychiatry, Psychology and Neuroscience, 4th floor New Hunt's House, Guy's Campus, London, UK
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London, E4 1NS, UK
| | - Richard J T Wingate
- MRC Centre for Neurodevelopmental Disorders, King's College London, Institute of Psychiatry, Psychology and Neuroscience, 4th floor New Hunt's House, Guy's Campus, London, UK.
| |
Collapse
|
7
|
Demirbilek H, Hatipoglu N, Gul U, Tatli ZU, Ellard S, Flanagan SE, De Franco E, Kurtoglu S. Permanent neonatal diabetes mellitus and neurological abnormalities due to a novel homozygous missense mutation in NEUROD1. Pediatr Diabetes 2018. [PMID: 29521454 DOI: 10.1111/pedi.12669] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The basic helix-loop-helix (bHLH) transcription factor, neuronal differentiation 1 (NEUROD1) (also known as BETA2) is involved in the development of neural elements and endocrine pancreas. Less than 10 reports of adult-onset non-insulin-dependent diabetes mellitus (NIDDM) due to heterozygous NEUROD1 mutations and 2 cases with permanent neonatal diabetes mellitus (PNDM) and neurological abnormalities due to homozygous NEUROD1 mutations have been published. A 13 year-old female was referred to endocrine department due to hyperglycemia. She was on insulin therapy following a diagnosis of neonatal diabetes mellitus (NDM) at the age of 9-weeks but missed regular follow-up. Parents are second cousin. There was a significant family history of adult onset NIDDM including patient's father. Auxological measurements were within normal ranges. On laboratory examination blood glucose was 33.2 mmol/L with undetectable c-peptide and glycosylated hemoglobin level of 8.9% (73.8 mmol/mol). She had developed difficulty in walking at the age of 4 years which had worsened over time. On further evaluation, a diagnosis of visual impairment, mental retardation, ataxic gait, retinitis pigmentosa and sensory-neural deafness were considered. Cranial magnetic resonance imaging revealed cerebellar hypoplasia. Molecular genetic analysis using targeted next generation sequencing detected a novel homozygous missense mutation, p.Ile150Asn(c.449T>A), in NEUROD1. Both parents and 2 unaffected siblings were heterozygous for the mutation. We report the third case of PNDM with neurological abnormalities caused by homozygous NEUROD1 mutation, the first caused by a missense mutation. Heterozygous carriers of the p.Ile150Asn mutation were either unaffected or diagnosed with diabetes in adulthood. It is currently unclear whether the NEUROD1 heterozygous mutation has contributed to diabetes development in these individuals.
Collapse
Affiliation(s)
- Huseyin Demirbilek
- Department of Paediatric Endocrinology, Hacettepe University Medical Faculty, Ankara, Turkey
| | - Nihal Hatipoglu
- Department of Paediatric Endocrinology, Erciyes University Medical Faculty, Kayseri, Turkey
| | - Ulku Gul
- Department of Paediatric Endocrinology, Erciyes University Medical Faculty, Kayseri, Turkey
| | - Zeynep U Tatli
- Department of Paediatric Endocrinology, Erciyes University Medical Faculty, Kayseri, Turkey
| | - Sian Ellard
- Department of Molecular Genetics, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Sarah E Flanagan
- Department of Molecular Genetics, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Elisa De Franco
- Department of Molecular Genetics, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Selim Kurtoglu
- Department of Paediatric Endocrinology, Erciyes University Medical Faculty, Kayseri, Turkey
| |
Collapse
|
8
|
Schilling K. Moving into shape: cell migration during the development and histogenesis of the cerebellum. Histochem Cell Biol 2018; 150:13-36. [DOI: 10.1007/s00418-018-1677-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2018] [Indexed: 12/31/2022]
|
9
|
Quezada-Ramírez M, Castañeda-Arellano R, Pérez-Sánchez G, Hernández-Soto J, Segovia J. The Growth arrest specific 1 ( Gas1 ) gene is transcriptionally regulated by NeuroD1 via two distal E-boxes. Exp Cell Res 2018; 363:332-341. [DOI: 10.1016/j.yexcr.2018.01.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 01/23/2018] [Accepted: 01/24/2018] [Indexed: 12/13/2022]
|
10
|
Feng W, Kawauchi D, Körkel-Qu H, Deng H, Serger E, Sieber L, Lieberman JA, Jimeno-González S, Lambo S, Hanna BS, Harim Y, Jansen M, Neuerburg A, Friesen O, Zuckermann M, Rajendran V, Gronych J, Ayrault O, Korshunov A, Jones DTW, Kool M, Northcott PA, Lichter P, Cortés-Ledesma F, Pfister SM, Liu HK. Chd7 is indispensable for mammalian brain development through activation of a neuronal differentiation programme. Nat Commun 2017; 8:14758. [PMID: 28317875 PMCID: PMC5364396 DOI: 10.1038/ncomms14758] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 01/25/2017] [Indexed: 12/16/2022] Open
Abstract
Mutations in chromatin modifier genes are frequently associated with neurodevelopmental diseases. We herein demonstrate that the chromodomain helicase DNA-binding protein 7 (Chd7), frequently associated with CHARGE syndrome, is indispensable for normal cerebellar development. Genetic inactivation of Chd7 in cerebellar granule neuron progenitors leads to cerebellar hypoplasia in mice, due to the impairment of granule neuron differentiation, induction of apoptosis and abnormal localization of Purkinje cells, which closely recapitulates known clinical features in the cerebella of CHARGE patients. Combinatory molecular analyses reveal that Chd7 is required for the maintenance of open chromatin and thus activation of genes essential for granule neuron differentiation. We further demonstrate that both Chd7 and Top2b are necessary for the transcription of a set of long neuronal genes in cerebellar granule neurons. Altogether, our comprehensive analyses reveal a mechanism with chromatin remodellers governing brain development via controlling a core transcriptional programme for cell-specific differentiation.
Collapse
Affiliation(s)
- Weijun Feng
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ–ZMBH Alliance, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Daisuke Kawauchi
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Huiqin Körkel-Qu
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ–ZMBH Alliance, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Huan Deng
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ–ZMBH Alliance, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Elisabeth Serger
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ–ZMBH Alliance, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Laura Sieber
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Jenna Ariel Lieberman
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, Sevilla 41092, Spain
| | - Silvia Jimeno-González
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, Sevilla 41092, Spain
| | - Sander Lambo
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Bola S. Hanna
- Molecular Genetics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Yassin Harim
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ–ZMBH Alliance, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Malin Jansen
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ–ZMBH Alliance, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Anna Neuerburg
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ–ZMBH Alliance, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Olga Friesen
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ–ZMBH Alliance, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Marc Zuckermann
- Molecular Genetics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Vijayanad Rajendran
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Jan Gronych
- Molecular Genetics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Olivier Ayrault
- Institut Curie, CNRS UMR 3347, INSERM U1021, Centre Universitaire, Bâtiment 110, 91405 Orsay, France
| | - Andrey Korshunov
- Clinical Cooperation Unit Neuropathology, German Cancer Research Centre (DKFZ), Department of Neuropathology, University of Heidelberg, Heidelberg 69120, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg 69120, Germany
| | - David T. W. Jones
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg 69120, Germany
| | - Marcel Kool
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Paul A. Northcott
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, USA
| | - Peter Lichter
- Molecular Genetics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg 69120, Germany
| | - Felipe Cortés-Ledesma
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, Sevilla 41092, Spain
| | - Stefan M. Pfister
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg 69120, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Hai-Kun Liu
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ–ZMBH Alliance, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| |
Collapse
|
11
|
Early Purkinje Cell Development and the Origins of Cerebellar Patterning. CONTEMPORARY CLINICAL NEUROSCIENCE 2017. [DOI: 10.1007/978-3-319-59749-2_4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
|
12
|
Tiberi L, Bonnefont J, van den Ameele J, Le Bon SD, Herpoel A, Bilheu A, Baron BW, Vanderhaeghen P. A BCL6/BCOR/SIRT1 complex triggers neurogenesis and suppresses medulloblastoma by repressing Sonic Hedgehog signaling. Cancer Cell 2014; 26:797-812. [PMID: 25490446 DOI: 10.1016/j.ccell.2014.10.021] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 07/27/2014] [Accepted: 10/30/2014] [Indexed: 12/21/2022]
Abstract
Disrupted differentiation during development can lead to oncogenesis, but the underlying mechanisms remain poorly understood. Here we identify BCL6, a transcriptional repressor and lymphoma oncoprotein, as a pivotal factor required for neurogenesis and tumor suppression of medulloblastoma (MB). BCL6 is necessary for and capable of preventing the development of GNP-derived MB in mice, and can block the growth of human MB cells in vitro. BCL6 neurogenic and oncosuppressor effects rely on direct transcriptional repression of Gli1 and Gli2 effectors of the SHH pathway, through recruitment of BCOR corepressor and SIRT1 deacetylase. Our findings identify the BCL6/BCOR/SIRT1 complex as a potent repressor of the SHH pathway in normal and oncogenic neural development, with direct diagnostic and/or therapeutic relevance for SHH MB.
Collapse
Affiliation(s)
- Luca Tiberi
- Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), and ULB Institute of Neuroscience (UNI), 808 Route de Lennik, 1070 Brussels, Belgium
| | - Jérôme Bonnefont
- Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), and ULB Institute of Neuroscience (UNI), 808 Route de Lennik, 1070 Brussels, Belgium
| | - Jelle van den Ameele
- Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), and ULB Institute of Neuroscience (UNI), 808 Route de Lennik, 1070 Brussels, Belgium
| | - Serge-Daniel Le Bon
- Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), and ULB Institute of Neuroscience (UNI), 808 Route de Lennik, 1070 Brussels, Belgium
| | - Adèle Herpoel
- Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), and ULB Institute of Neuroscience (UNI), 808 Route de Lennik, 1070 Brussels, Belgium
| | - Angéline Bilheu
- Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), and ULB Institute of Neuroscience (UNI), 808 Route de Lennik, 1070 Brussels, Belgium
| | - Beverly W Baron
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Pierre Vanderhaeghen
- Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), and ULB Institute of Neuroscience (UNI), 808 Route de Lennik, 1070 Brussels, Belgium; Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Université Libre de Bruxelles, 808 Route de Lennik, 1070 Brussels, Belgium.
| |
Collapse
|
13
|
Benitez SG, Castro AE, Patterson SI, Muñoz EM, Seltzer AM. Hypoxic preconditioning differentially affects GABAergic and glutamatergic neuronal cells in the injured cerebellum of the neonatal rat. PLoS One 2014; 9:e102056. [PMID: 25032984 PMCID: PMC4102512 DOI: 10.1371/journal.pone.0102056] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 06/13/2014] [Indexed: 12/13/2022] Open
Abstract
In this study we examined cerebellar alterations in a neonatal rat model of hypoxic-ischemic brain injury with or without hypoxic preconditioning (Pc). Between postnatal days 7 and 15, the cerebellum is still undergoing intense cellular proliferation, differentiation and migration, dendritogenesis and synaptogenesis. The expression of glutamate decarboxylase 1 (GAD67) and the differentiation factor NeuroD1 were examined as markers of Purkinje and granule cells, respectively. We applied quantitative immunohistochemistry to sagittal cerebellar slices, and Western blot analysis of whole cerebella obtained from control (C) rats and rats submitted to Pc, hypoxia-ischemia (L) and a combination of both treatments (PcL). We found that either hypoxia-ischemia or Pc perturbed the granule cells in the posterior lobes, affecting their migration and final placement in the internal granular layer. These effects were partially attenuated when the Pc was delivered prior to the hypoxia-ischemia. Interestingly, whole nuclear NeuroD1 levels in Pc animals were comparable to those in the C rats. However, a subset of Purkinje cells that were severely affected by the hypoxic-ischemic insult—showing signs of neuronal distress at the levels of the nucleus, cytoplasm and dendritic arborization—were not protected by Pc. A monoclonal antibody specific for GAD67 revealed a three-band pattern in cytoplasmic extracts from whole P15 cerebella. A ∼110 kDa band, interpreted as a potential homodimer of a truncated form of GAD67, was reduced in Pc and L groups while its levels were close to the control animals in PcL rats. Additionally we demonstrated differential glial responses depending on the treatment, including astrogliosis in hypoxiated cerebella and a selective effect of hypoxia-ischemia on the vimentin-immunolabeled intermediate filaments of the Bergmann glia. Thus, while both glutamatergic and GABAergic cerebellar neurons are compromised by the hypoxic-ischemic insult, the former are protected by a preconditioning hypoxia while the latter are not.
Collapse
Affiliation(s)
- Sergio G Benitez
- Laboratory of Neurobiology: Chronobiology Section, Institute of Histology and Embryology of Mendoza (IHEM), School of Medicine, National University of Cuyo, Mendoza, National Scientific and Technical Research Council (CONICET), National Agency for Scientific and Technological Promotion (ANPCyT), Mendoza, Argentina
| | - Analía E Castro
- Laboratory of Neurobiology: Chronobiology Section, Institute of Histology and Embryology of Mendoza (IHEM), School of Medicine, National University of Cuyo, Mendoza, National Scientific and Technical Research Council (CONICET), National Agency for Scientific and Technological Promotion (ANPCyT), Mendoza, Argentina
| | - Sean I Patterson
- Traumatic and Toxic Lesions in the Nervous System Section, Institute of Histology and Embryology of Mendoza (IHEM), School of Medicine, National University of Cuyo, Mendoza, National Scientific and Technical Research Council (CONICET), National Agency for Scientific and Technological Promotion (ANPCyT), Mendoza, Argentina
| | - Estela M Muñoz
- Laboratory of Neurobiology: Chronobiology Section, Institute of Histology and Embryology of Mendoza (IHEM), School of Medicine, National University of Cuyo, Mendoza, National Scientific and Technical Research Council (CONICET), National Agency for Scientific and Technological Promotion (ANPCyT), Mendoza, Argentina
| | - Alicia M Seltzer
- Neonatal Brain Development Section, Institute of Histology and Embryology of Mendoza (IHEM), School of Medicine, National University of Cuyo, Mendoza, National Scientific and Technical Research Council (CONICET), National Agency for Scientific and Technological Promotion (ANPCyT), Mendoza, Argentina
| |
Collapse
|
14
|
Alvarez-Saavedra M, De Repentigny Y, Lagali PS, Raghu Ram EVS, Yan K, Hashem E, Ivanochko D, Huh MS, Yang D, Mears AJ, Todd MAM, Corcoran CP, Bassett EA, Tokarew NJA, Kokavec J, Majumder R, Ioshikhes I, Wallace VA, Kothary R, Meshorer E, Stopka T, Skoultchi AI, Picketts DJ. Snf2h-mediated chromatin organization and histone H1 dynamics govern cerebellar morphogenesis and neural maturation. Nat Commun 2014; 5:4181. [PMID: 24946904 PMCID: PMC4083431 DOI: 10.1038/ncomms5181] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 05/15/2014] [Indexed: 12/28/2022] Open
Abstract
Chromatin compaction mediates progenitor to post-mitotic cell transitions and modulates gene expression programs, yet the mechanisms are poorly defined. Snf2h and Snf2l are ATP-dependent chromatin remodelling proteins that assemble, reposition and space nucleosomes, and are robustly expressed in the brain. Here we show that mice conditionally inactivated for Snf2h in neural progenitors have reduced levels of histone H1 and H2A variants that compromise chromatin fluidity and transcriptional programs within the developing cerebellum. Disorganized chromatin limits Purkinje and granule neuron progenitor expansion, resulting in abnormal post-natal foliation, while deregulated transcriptional programs contribute to altered neural maturation, motor dysfunction and death. However, mice survive to young adulthood, in part from Snf2l compensation that restores Engrailed-1 expression. Similarly, Purkinje-specific Snf2h ablation affects chromatin ultrastructure and dendritic arborization, but alters cognitive skills rather than motor control. Our studies reveal that Snf2h controls chromatin organization and histone H1 dynamics for the establishment of gene expression programs underlying cerebellar morphogenesis and neural maturation.
Collapse
Affiliation(s)
- Matías Alvarez-Saavedra
- 1] Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6 [2] Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| | - Yves De Repentigny
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6
| | - Pamela S Lagali
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6
| | - Edupuganti V S Raghu Ram
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Keqin Yan
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6
| | - Emile Hashem
- 1] Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6 [2] Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| | - Danton Ivanochko
- 1] Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6 [2] Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| | - Michael S Huh
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6
| | - Doo Yang
- 1] Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5 [2] Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| | - Alan J Mears
- Vision Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6
| | - Matthew A M Todd
- 1] Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6 [2] Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| | - Chelsea P Corcoran
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6
| | - Erin A Bassett
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| | - Nicholas J A Tokarew
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| | - Juraj Kokavec
- Institute of Pathologic Physiology, First Faculty of Medicine, Charles University in Prague, Prague 12853, Czech Republic
| | - Romit Majumder
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Ilya Ioshikhes
- 1] Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5 [2] Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| | - Valerie A Wallace
- 1] Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5 [2] Vision Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6
| | - Rashmi Kothary
- 1] Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6 [2] Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| | - Eran Meshorer
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Tomas Stopka
- Institute of Pathologic Physiology, First Faculty of Medicine, Charles University in Prague, Prague 12853, Czech Republic
| | - Arthur I Skoultchi
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - David J Picketts
- 1] Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6 [2] Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5 [3] Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| |
Collapse
|
15
|
Kim BJ, Scott DA. Mouse model reveals the role of RERE in cerebellar foliation and the migration and maturation of Purkinje cells. PLoS One 2014; 9:e87518. [PMID: 24466353 PMCID: PMC3900724 DOI: 10.1371/journal.pone.0087518] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 12/22/2013] [Indexed: 11/22/2022] Open
Abstract
Nuclear receptors and their coregulators play a critical role in brain development by regulating the spatiotemporal expression of their target genes. The arginine-glutamic acid dipeptide repeats gene (Rere) encodes a nuclear receptor coregulator previously known as Atrophin 2. In the developing cerebellum, RERE is expressed in the molecular layer, the Purkinje cell layer and the granule cell layer but not in granule cell precursors. To study RERE's role in cerebellar development, we used RERE-deficient embryos bearing a null allele (om) and a hypomorphic allele (eyes3) of Rere (Rereom/eyes3). In contrast to wild-type embryos, formation of the principal fissures in these RERE-deficient embryos was delayed and the proliferative activity of granule cell precursors (GCPs) was reduced at E18.5. This reduction in proliferation was accompanied by a decrease in the expression of sonic hedgehog (SHH), which is secreted from Purkinje cells and is required for normal GCP proliferation. The maturation and migration of Purkinje cells in Rereom/eyes3 embryos was also delayed with decreased numbers of post-migratory Purkinje cells in the cerebellum. During the postnatal period, RERE depletion caused incomplete division of lobules I/II and III due to truncated development of the precentral fissure in the cerebellar vermis, abnormal development of lobule crus I and lobule crus II in the cerebellar hemispheres due to attenuation of the intercrural fissure, and decreased levels of Purkinje cell dendritic branching. We conclude that RERE-deficiency leads to delayed development of the principal fissures and delayed maturation and migration of Purkinje cells during prenatal cerebellar development and abnormal cerebellar foliation and Purkinje cell maturation during postnatal cerebellar development.
Collapse
Affiliation(s)
- Bum Jun Kim
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Daryl A. Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
16
|
D'Amico LA, Boujard D, Coumailleau P. The neurogenic factor NeuroD1 is expressed in post-mitotic cells during juvenile and adult Xenopus neurogenesis and not in progenitor or radial glial cells. PLoS One 2013; 8:e66487. [PMID: 23799108 PMCID: PMC3683004 DOI: 10.1371/journal.pone.0066487] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Accepted: 05/07/2013] [Indexed: 12/21/2022] Open
Abstract
In contrast to mammals that have limited proliferation and neurogenesis capacities, the Xenopus frog exhibit a great potential regarding proliferation and production of new cells in the adult brain. This ability makes Xenopus a useful model for understanding the molecular programs required for adult neurogenesis. Transcriptional factors that control adult neurogenesis in vertebrate species undergoing widespread neurogenesis are unknown. NeuroD1 is a member of the family of proneural genes, which function during embryonic neurogenesis as a potent neuronal differentiation factor. Here, we study in detail the expression of NeuroD1 gene in the juvenile and adult Xenopus brains by in situ hybridization combined with immunodetections for proliferation markers (PCNA, BrdU) or in situ hybridizations for cell type markers (Vimentin, Sox2). We found NeuroD1 gene activity in many brain regions, including olfactory bulbs, pallial regions of cerebral hemispheres, preoptic area, habenula, hypothalamus, cerebellum and medulla oblongata. We also demonstrated by double staining NeuroD1/BrdU experiments, after long post-BrdU administration survival times, that NeuroD1 gene activity was turned on in new born neurons during post-metamorphic neurogenesis. Importantly, we provided evidence that NeuroD1-expressing cells at this brain developmental stage were post-mitotic (PCNA-) cells and not radial glial (Vimentin+) or progenitors (Sox2+) cells.
Collapse
Affiliation(s)
- Laure Anne D'Amico
- Centre de Ressources Biologiques Xénope UMS U3387, CNRS, and Rennes 1 University, Rennes, France
| | - Daniel Boujard
- Centre de Ressources Biologiques Xénope UMS U3387, CNRS, and Rennes 1 University, Rennes, France
| | - Pascal Coumailleau
- Centre de Ressources Biologiques Xénope UMS U3387, CNRS, and Rennes 1 University, Rennes, France
- IRSET U1085, INSERM and Rennes1 University, Rennes, France
- * E-mail:
| |
Collapse
|
17
|
Mandal NA, Tran JTA, Saadi A, Rahman AK, Huynh TP, Klein WH, Cho JH. Expression and localization of CERKL in the mammalian retina, its response to light-stress, and relationship with NeuroD1 gene. Exp Eye Res 2012; 106:24-33. [PMID: 23142158 DOI: 10.1016/j.exer.2012.10.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 10/25/2012] [Accepted: 10/26/2012] [Indexed: 01/30/2023]
Abstract
Mutations in the Ceramide kinase like (CERKL) gene are associated with retinitis pigmentosa (RP26) and cone-rod dystrophy. CERKL is homologous to Ceramide kinase (CERK), and its function is still unknown. The purpose of this study was to test the expression and distribution of this gene and its protein in rat and in mouse tissues, in light-stressed rat retinas and in the retinas of NeuroD1 knock-out mice to understand the role of CERKL in the retina. Expression of Cerkl and Cerk mRNA was determined by quantitative RT-PCR. Expression of the protein was determined by Western blotting with anti-CERKL antibody. Localization of the protein was determined by using immunofluorescence microscopy. With qRT-PCR, we revealed that the relative mRNA expression of Cerkl was the highest in the retina among all the rat tissue tested; it was >10-fold higher than in the brain. On the other hand, Cerk has ubiquitous expression and its relative abundance is >2 fold of Cerkl in the retina. Cerkl was expressed minimally in the developing mouse eyes and reached a peak at retinal maturity at 2 months. Western blots of retinal tissues revealed two major CERKL protein bands: 59 kDa (C1) and 37 kDa (C2). However, only C2 CERKL was found in the rat retinal rod outer segment (ROS) at level of that was not changed in light vs. dark adaptation. In the light-stressed retina, expression of Cerkl mRNA increased significantly, which was reflected in only on C2 CERKL protein. The CERKL protein localized prominently to the ganglion cells, inner nuclear layers (INL), retinal pigment epithelial (RPE) cells, and photoreceptor inner segments in the retinal sections. Nuclear localization of CERKL was not affected in RPE, INL and the ganglion cell layers in the light-stressed retina; however, the perinuclear and outer segment locations appear to be altered. In the NeuroD1 knock-out mouse retina, the expression of Cerkl mRNA and protein decreased and that decrease also pertains to C2 CERKL. In conclusion, the retina had the highest level of Cerkl mRNA and protein expression, which reached its maximum in the adult retina; CERKL localized to ROS and RPE cells and the light-adaptation did not change the level of CERKL in ROS; light-stress induced Cerkl expression in the retina; and its expression decreased in NeuroD1 knock-out retina. Thus, CERKL may be important for the stress responses and protection of photoreceptor cells.
Collapse
Affiliation(s)
- Nawajes A Mandal
- Department of Ophthalmology, OUHSC, Dean A. McGee Eye Institute, Oklahoma City, OK 73104, United States.
| | | | | | | | | | | | | |
Collapse
|
18
|
Fukuoka T, Sumida K, Yamada T, Higuchi C, Nakagaki K, Nakamura K, Kohsaka S, Saito K, Oeda K. Gene expression profiles in the common marmoset brain determined using a newly developed common marmoset-specific DNA microarray. Neurosci Res 2010; 66:62-85. [DOI: 10.1016/j.neures.2009.09.1709] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Revised: 08/28/2009] [Accepted: 09/28/2009] [Indexed: 10/20/2022]
|
19
|
Pan N, Jahan I, Lee JE, Fritzsch B. Defects in the cerebella of conditional Neurod1 null mice correlate with effective Tg(Atoh1-cre) recombination and granule cell requirements for Neurod1 for differentiation. Cell Tissue Res 2009; 337:407-28. [PMID: 19609565 DOI: 10.1007/s00441-009-0826-6] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2009] [Accepted: 06/12/2009] [Indexed: 01/19/2023]
Abstract
Neurod1 is a crucial basic helix-loop-helix gene for most cerebellar granule cells and mediates the differentiation of these cells downstream of Atoh1-mediated proliferation of the precursors. In Neurod1 null mice, granule cells die throughout the posterior two thirds of the cerebellar cortex during development. However, Neurod1 is also necessary for pancreatic beta-cell development, and therefore Neurod1 null mice are diabetic, which potentially influences cerebellar defects. Here, we report a new Neurod1 conditional knock-out mouse model created by using a Tg(Atoh1-cre) line to eliminate Neurod1 in the cerebellar granule cell precursors. Our data confirm and extend previous work on systemic Neurod1 null mice and show that, in the central lobules, granule cells can be eradicated in the absence of Neurod1. Granule cells in the anterior lobules are partially viable and depend on as yet unknown genes, but the Purkinje cells show defects not previously recognized. Interestingly, delayed and incomplete Tg(Atoh1-cre) upregulation occurs in the most posterior lobules; this leads to near normal expression of Neurod1 with a concomitant normal differentiation of granule cells, Purkinje cells, and unipolar brush cells in lobules IX and X. Our analysis suggests that Neurod1 negatively regulates Atoh1 to ensure a rapid transition from proliferative precursors to differentiating neurons. Our data have implications for research on medulloblastoma, one of the most frequent brain tumors of children, as the results suggest that targeted overexpression of Neurod1 under Atoh1 promoter control may initiate the differentiation of these tumors.
Collapse
Affiliation(s)
- Ning Pan
- Department of Biology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
20
|
Abstract
Thyroid hormone (TH) plays a key role in mammalian brain development. The developing brain is sensitive to both TH deficiency and excess. Brain development in the absence of TH results in motor skill deficiencies and reduced intellectual development. These functional abnormalities can be attributed to maldevelopment of specific cell types and regions of the brain including the cerebellum. TH functions at the molecular level by regulating gene transcription. Therefore, understanding how TH regulates cerebellar development requires identification of TH-regulated gene targets and the cells expressing these genes. Additionally, the process of TH-dependent regulation of gene expression is tightly controlled by mechanisms including regulation of TH transport, TH metabolism, toxicologic inhibition of TH signaling, and control of the nuclear TH response apparatus. This review will describe the functional, cellular, and molecular effects of TH deficit in the developing cerebellum and emphasize the most recent findings regarding TH action in this important brain region.
Collapse
Affiliation(s)
- Grant W Anderson
- Department of Pharmacy Practice and Pharmaceutical Sciences, College of Pharmacy, University of Minnesota, Duluth, Minnesota 55812, USA.
| |
Collapse
|
21
|
Kim BJ, Takamoto N, Yan J, Tsai SY, Tsai MJ. Chicken Ovalbumin Upstream Promoter-Transcription Factor II (COUP-TFII) regulates growth and patterning of the postnatal mouse cerebellum. Dev Biol 2008; 326:378-91. [PMID: 19041640 DOI: 10.1016/j.ydbio.2008.11.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2008] [Revised: 10/29/2008] [Accepted: 11/03/2008] [Indexed: 01/26/2023]
Abstract
COUP-TFII (also known as Nr2f2), a member of the nuclear orphan receptor superfamily, is expressed in several regions of the central nervous system (CNS), including the ventral thalamus, hypothalamus, midbrain, pons, and spinal cord. To address the function of COUP-TFII in the CNS, we generated conditional COUP-TFII knockout mice using a tissue-specific NSE-Cre recombinase. Ablation of COUP-TFII in the brain resulted in malformation of the lobule VI in the cerebellum and a decrease in differentiation of cerebellar neurons and cerebellar growth. The decrease in cerebellar growth in NSE(Cre/+)/CII(F/F) mice is due to reduced proliferation and increased apoptosis in granule cell precursors (GCPs). Additional studies demonstrated that insulin like growth factor 1 (IGF-1) expression was reduced in the cerebellum of NSE(Cre/+)/CII(F/F) mice, thereby leading to decreased Akt1 and GSK-3beta activities, and the reduced expression of mTOR. Using ChIP assays, we demonstrated that COUP-TFII was recruited to the promoter region of IGF-1 in a Sp1-dependent manner. In addition, dendritic branching of Purkinje cells was decreased in the mutant mice. Thus, our results indicate that COUP-TFII regulates growth and maturation of the mouse postnatal cerebellum through modulation of IGF-1 expression.
Collapse
Affiliation(s)
- Bum Jun Kim
- Department of Molecular and Cellular Biology and Development Program, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
22
|
Abstract
The thioredoxin-dependent system is an essential regulator of cellular redox balance. Since oxidative stress has been linked with neurodegenerative disease, we studied the roles of thioredoxin reductases in brain using mice with nervous system (NS)-specific deletion of cytosolic (Txnrd1) and mitochondrial (Txnrd2) thioredoxin reductase. While NS-specific Txnrd2 null mice develop normally, mice lacking Txnrd1 in the NS were significantly smaller and displayed ataxia and tremor. A striking patterned cerebellar hypoplasia was observed. Proliferation of the external granular layer (EGL) was strongly reduced and fissure formation and laminar organisation of the cerebellar cortex was impaired in the rostral portion of the cerebellum. Purkinje cells were ectopically located and their dendrites stunted. The Bergmann glial network was disorganized and showed a pronounced reduction in fiber strength. Cerebellar hypoplasia did not result from increased apoptosis, but from decreased proliferation of granule cell precursors within the EGL. Of note, neuron-specific inactivation of Txnrd1 did not result in cerebellar hypoplasia, suggesting a vital role for Txnrd1 in Bergmann glia or neuronal precursor cells.
Collapse
|
23
|
Neptune ER, Podowski M, Calvi C, Cho JH, Garcia JGN, Tuder R, Linnoila RI, Tsai MJ, Dietz HC. Targeted disruption of NeuroD, a proneural basic helix-loop-helix factor, impairs distal lung formation and neuroendocrine morphology in the neonatal lung. J Biol Chem 2008; 283:21160-9. [PMID: 18339630 PMCID: PMC2475704 DOI: 10.1074/jbc.m708692200] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Despite the importance of airspace integrity in vertebrate gas exchange,
the molecular pathways that instruct distal lung formation are poorly
understood. Recently, we found that fibrillin-1 deficiency in mice impairs
alveolar formation and recapitulates the pulmonary features of human Marfan
syndrome. To further elucidate effectors involved in distal lung formation, we
performed expression profiling analysis comparing the fibrillin-1-deficient
and wild-type developing lung. NeuroD, a basic helix-loop-helix transcription
factor, fulfilled the expression criteria for a candidate mediator of distal
lung development. We investigated its role in murine lung development using
genetically targeted NeuroD-deficient mice. We found that NeuroD deficiency
results in both impaired alveolar septation and altered morphology of the
pulmonary neuroendocrine cells. NeuroD-deficient mice had enlarged alveoli
associated with reduced epithelial proliferation in the airway and airspace
compartments during development. Additionally, the neuroendocrine compartment
in these mice manifested an increased number of neuroepithelial bodies but a
reduced number of solitary pulmonary neuroendocrine cells in the neonatal
lung. Overexpression of NeuroD in a murine lung epithelial cell line conferred
a neuroendocrine phenotype characterized by the induction of neuroendocrine
markers as well as increased proliferation. These results support an
unanticipated role for NeuroD in the regulation of pulmonary neuroendocrine
and alveolar morphogenesis and suggest an intimate connection between the
neuroendocrine compartment and distal lung development.
Collapse
Affiliation(s)
- Enid R Neptune
- Division of Pulmonary and Critical Care Medicine, Institute of Genetic Medicine, Howard Hughes Medical Institute, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Qin J, Suh JM, Kim BJ, Yu CT, Tanaka T, Kodama T, Tsai MJ, Tsai SY. The expression pattern of nuclear receptors during cerebellar development. Dev Dyn 2007; 236:810-20. [PMID: 17205580 DOI: 10.1002/dvdy.21060] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The cerebellum is essential for fine control of movement and posture, and it has been a useful model for studying many aspects of neural development because of its relatively simple anatomy and developmental program. However, the roles of nuclear receptors (NRs) underlying formation of the cerebellum and maintenance of cerebellar functions are still poorly characterized. As a contribution to the Nuclear Receptor Signaling Atlas (NURSA), we employed immunohistochemistry to investigate the expression pattern of 18 NRs in the cerebellum. Ten receptors were demonstrated to be expressed in the postnatal day 21 (P21) cerebellum. Among them, five receptors (COUP-TFI, COUP-TFII, RORalpha, ERbeta, and ERRgamma) were expressed at all stages (embryonic stage, P0, P7, and P21) examined. Interestingly, COUP-TFI and COUP-TFII show differential anterior-posterior expression patterns during cerebellar development. Taken together, our results suggest that members of the nuclear receptor superfamily might play importantly physiological roles in the cerebellum.
Collapse
Affiliation(s)
- Jun Qin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Cho JH, Klein WH, Tsai MJ. Compensational regulation of bHLH transcription factors in the postnatal development of BETA2/NeuroD1-null retina. Mech Dev 2007; 124:543-50. [PMID: 17629466 PMCID: PMC4300853 DOI: 10.1016/j.mod.2007.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2007] [Revised: 05/24/2007] [Accepted: 06/02/2007] [Indexed: 10/23/2022]
Abstract
The bHLH transcriptional factor BETA2/NeuroD1 is essential for the survival of photoreceptor cells in the retina. Although this gene is expressed throughout the retina, BETA2/NeuroD1 knockout mice show photoreceptor cell degeneration only in the outer nuclear layer of the retina; other retinal neurons are not affected. Previous studies on retina explants lacking three bHLH genes revealed that retinal neurons in the inner nuclear layer require multiple bHLH genes for their differentiation and survival. However, single- or double-gene mutations show no or a lesser degree of abnormalities during eye development, likely because of compensation or cooperative regulation among those genes. Because not all null mice survive until the retina is fully organized, no direct evidence of this concept has been reported. To understand the regulatory mechanisms between bHLH factors in retinal development, we performed a detailed analysis of BETA2/NeuroD1 knockout mice. BETA2/NeuroD1 was expressed in all 3 layers of the mouse retina, including all major types of neurons. In addition, a null mutation of BETA2/NeuroD1 resulted in up-regulation of other bHLH genes, Mash1, Neurogenin2, and Math3, in the inner nuclear layer. Our data suggest that compensatory and cross regulatory mechanisms exist among the bHLH factors during retinal development.
Collapse
Affiliation(s)
- Jang-Hyeon Cho
- Department of Biochemistry and Molecular Biology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Blvd. Unit 1000, Houston, TX 77030, USA.
| | | | | |
Collapse
|