1
|
Perycz M, Dabrowski MJ, Jardanowska-Kotuniak M, Roura AJ, Gielniewski B, Stepniak K, Dramiński M, Ciechomska IA, Kaminska B, Wojtas B. Comprehensive analysis of the REST transcription factor regulatory networks in IDH mutant and IDH wild-type glioma cell lines and tumors. Acta Neuropathol Commun 2024; 12:72. [PMID: 38711090 PMCID: PMC11071216 DOI: 10.1186/s40478-024-01779-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 04/09/2024] [Indexed: 05/08/2024] Open
Abstract
The RE1-silencing transcription factor (REST) acts either as a repressor or activator of transcription depending on the genomic and cellular context. REST is a key player in brain cell differentiation by inducing chromatin modifications, including DNA methylation, in a proximity of its binding sites. Its dysfunction may contribute to oncogenesis. Mutations in IDH1/2 significantly change the epigenome contributing to blockade of cell differentiation and glioma development. We aimed at defining how REST modulates gene activation and repression in the context of the IDH mutation-related phenotype in gliomas. We studied the effects of REST knockdown, genome wide occurrence of REST binding sites, and DNA methylation of REST motifs in IDH wild type and IDH mutant gliomas. We found that REST target genes, REST binding patterns, and TF motif occurrence proximal to REST binding sites differed in IDH wild-type and mutant gliomas. Among differentially expressed REST targets were genes involved in glial cell differentiation and extracellular matrix organization, some of which were differentially methylated at promoters or gene bodies. REST knockdown differently impacted invasion of the parental or IDH1 mutant glioma cells. The canonical REST-repressed gene targets showed significant correlation with the GBM NPC-like cellular state. Interestingly, results of REST or KAISO silencing suggested the interplay between these TFs in regulation of REST-activated and repressed targets. The identified gene regulatory networks and putative REST cooperativity with other TFs, such as KAISO, show distinct REST target regulatory networks in IDH-WT and IDH-MUT gliomas, without concomitant DNA methylation changes. We conclude that REST could be an important therapeutic target in gliomas.
Collapse
Affiliation(s)
- Malgorzata Perycz
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | - Michal J Dabrowski
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | - Marta Jardanowska-Kotuniak
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
- Doctoral School of Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Adria-Jaume Roura
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Bartlomiej Gielniewski
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Karolina Stepniak
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Michał Dramiński
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | - Iwona A Ciechomska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Bartosz Wojtas
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.
- Laboratory of Sequencing, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
2
|
Talley MJ, Nardini D, Ehrman LA, Lu QR, Waclaw RR. Distinct requirements for Tcf3 and Tcf12 during oligodendrocyte development in the mouse telencephalon. Neural Dev 2023; 18:5. [PMID: 37684687 PMCID: PMC10485956 DOI: 10.1186/s13064-023-00173-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/31/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND E-proteins encoded by Tcf3, Tcf4, and Tcf12 are class I basic helix-loop-helix (bHLH) transcription factors (TFs) that are thought to be widely expressed during development. However, their function in the developing brain, specifically in the telencephalon remains an active area of research. Our study examines for the first time if combined loss of two E-proteins (Tcf3 and Tcf12) influence distinct cell fates and oligodendrocyte development in the mouse telencephalon. METHODS We generated Tcf3/12 double conditional knockouts (dcKOs) using Olig2Cre/+ or Olig1Cre/+ to overcome compensatory mechanisms between E-proteins and to understand the specific requirement for Tcf3 and Tcf12 in the ventral telencephalon and during oligodendrogenesis. We utilized a combination of in situ hybridization, immunohistochemistry, and immunofluorescence to address development of the telencephalon and oligodendrogenesis at embryonic and postnatal stages in Tcf3/12 dcKOs. RESULTS We show that the E-proteins Tcf3 and Tcf12 are expressed in progenitors of the embryonic telencephalon and throughout the oligodendrocyte lineage in the postnatal brain. Tcf3/12 dcKOs showed transient defects in progenitor cells with an enlarged medial ganglionic eminence (MGE) region which correlated with reduced generation of embryonic oligodendrocyte progenitor cells (OPCs) and increased expression of MGE interneuron genes. Postnatal Tcf3/12 dcKOs showed a recovery of OPCs but displayed a sustained reduction in mature oligodendrocytes (OLs). Interestingly, Tcf4 remained expressed in the dcKOs suggesting that it cannot compensate for the loss of Tcf3 and Tcf12. Generation of Tcf3/12 dcKOs with Olig1Cre/+ avoided the MGE morphology defect caused by Olig2Cre/+ but dcKOs still exhibited reduced embryonic OPCs and subsequent reduction in postnatal OLs. CONCLUSION Our data reveal that Tcf3 and Tcf12 play a role in controlling OPC versus cortical interneuron cell fate decisions in MGE progenitors in addition to playing roles in the generation of embryonic OPCs and differentiation of postnatal OLs in the oligodendrocyte lineage.
Collapse
Affiliation(s)
- Mary Jo Talley
- Graduate Program in Molecular and Developmental Biology, Cincinnati Children's Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Diana Nardini
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Lisa A Ehrman
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Q Richard Lu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Ronald R Waclaw
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA.
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.
| |
Collapse
|
3
|
Kang M, Yao Y. Laminin regulates oligodendrocyte development and myelination. Glia 2021; 70:414-429. [PMID: 34773273 DOI: 10.1002/glia.24117] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 11/08/2022]
Abstract
Oligodendrocytes are the cells that myelinate axons and provide trophic support to neurons in the CNS. Their dysfunction has been associated with a group of disorders known as demyelinating diseases, such as multiple sclerosis. Oligodendrocytes are derived from oligodendrocyte precursor cells, which differentiate into premyelinating oligodendrocytes and eventually mature oligodendrocytes. The development and function of oligodendrocytes are tightly regulated by a variety of molecules, including laminin, a major protein of the extracellular matrix. Accumulating evidence suggests that laminin actively regulates every aspect of oligodendrocyte biology, including survival, migration, proliferation, differentiation, and myelination. How can laminin exert such diverse functions in oligodendrocytes? It is speculated that the distinct laminin isoforms, laminin receptors, and/or key signaling molecules expressed in oligodendrocytes at different developmental stages are the reasons. Understanding molecular targets and signaling pathways unique to each aspect of oligodendrocyte biology will enable more accurate manipulation of oligodendrocyte development and function, which may have implications in the therapies of demyelinating diseases. Here in this review, we first introduce oligodendrocyte biology, followed by the expression of laminin and laminin receptors in oligodendrocytes and other CNS cells. Next, the functions of laminin in oligodendrocyte biology, including survival, migration, proliferation, differentiation, and myelination, are discussed in detail. Last, key questions and challenges in the field are discussed. By providing a comprehensive review on laminin's roles in OL lineage cells, we hope to stimulate novel hypotheses and encourage new research in the field.
Collapse
Affiliation(s)
- Minkyung Kang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Yao Yao
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
4
|
Zhang H, Younsi A, Zheng G, Tail M, Harms AK, Roth J, Hatami M, Skutella T, Unterberg A, Zweckberger K. Sonic Hedgehog modulates the inflammatory response and improves functional recovery after spinal cord injury in a thoracic contusion-compression model. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2021; 30:1509-1520. [PMID: 33704579 DOI: 10.1007/s00586-021-06796-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 01/15/2021] [Accepted: 02/24/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE The Sonic Hedgehog (Shh) pathway has been associated with a protective role after injury to the central nervous system (CNS). We, therefore, investigated the effects of intrathecal Shh-administration in the subacute phase after thoracic spinal cord injury (SCI) on secondary injury processes in rats. METHODS Twenty-one Wistar rats were subjected to thoracic clip-contusion/compression SCI at T9. Animals were randomized into three treatment groups (Shh, Vehicle, Sham). Seven days after SCI, osmotic pumps were implanted for seven-day continuous intrathecal administration of Shh. Basso, Beattie and Bresnahan (BBB) score, Gridwalk test and bodyweight were weekly assessed. Animals were sacrificed six weeks after SCI and immunohistological analyses were conducted. The results were compared between groups and statistical analysis was performed (p < 0.05 was considered significant). RESULTS The intrathecal administration of Shh led to significantly increased polarization of macrophages toward the anti-inflammatory M2-phenotype, significantly decreased T-lymphocytic invasion and significantly reduced resident microglia six weeks after the injury. Reactive astrogliosis was also significantly reduced while changes in size of the posttraumatic cyst as well as the overall macrophagic infiltration, although reduced, remained insignificant. Finally, with the administration of Shh, gain of bodyweight (216.6 ± 3.65 g vs. 230.4 ± 5.477 g; p = 0.0111) and BBB score (8.2 ± 0.2 vs. 5.9 ± 0.7 points; p = 0.0365) were significantly improved compared to untreated animals six weeks after SCI as well. CONCLUSION Intrathecal Shh-administration showed neuroprotective effects with attenuated neuroinflammation, reduced astrogliosis and improved functional recovery six weeks after severe contusion/compression SCI.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Neurosurgery, University Hospital Heidelberg, INF 400, 69120, Heidelberg, Germany
| | - Alexander Younsi
- Department of Neurosurgery, University Hospital Heidelberg, INF 400, 69120, Heidelberg, Germany.
| | - Guoli Zheng
- Department of Neurosurgery, University Hospital Heidelberg, INF 400, 69120, Heidelberg, Germany
| | - Mohamed Tail
- Department of Neurosurgery, University Hospital Heidelberg, INF 400, 69120, Heidelberg, Germany
| | - Anna-Kathrin Harms
- Department of Neurosurgery, University Hospital Heidelberg, INF 400, 69120, Heidelberg, Germany
| | - Judith Roth
- Department of Neurosurgery, University Hospital Heidelberg, INF 400, 69120, Heidelberg, Germany
| | - Maryam Hatami
- Department of Neuroanatomy, Institute for Anatomy and Cell Biology, University of Heidelberg, INF 307, 69120, Heidelberg, Germany
| | - Thomas Skutella
- Department of Neuroanatomy, Institute for Anatomy and Cell Biology, University of Heidelberg, INF 307, 69120, Heidelberg, Germany
| | - Andreas Unterberg
- Department of Neurosurgery, University Hospital Heidelberg, INF 400, 69120, Heidelberg, Germany
| | - Klaus Zweckberger
- Department of Neurosurgery, University Hospital Heidelberg, INF 400, 69120, Heidelberg, Germany
| |
Collapse
|
5
|
Gou X, Tang Y, Qu Y, Xiao D, Ying J, Mu D. Could the inhibitor of DNA binding 2 and 4 play a role in white matter injury? Rev Neurosci 2019; 30:625-638. [PMID: 30738015 DOI: 10.1515/revneuro-2018-0090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/02/2018] [Indexed: 01/12/2023]
Abstract
Abstract
White matter injury (WMI) prevents the normal development of myelination, leading to central nervous system myelination disorders and the production of chronic sequelae associated with WMI, such as chronic dyskinesia, cognitive impairment and cerebral palsy. This results in a large emotional and socioeconomic burden. Decreased myelination in preterm infant WMI is associated with the delayed development or destruction of oligodendrocyte (OL) lineage cells, particularly oligodendrocyte precursor cells (OPCs). The development of cells from the OL lineage involves the migration, proliferation and different stages of OL differentiation, finally leading to myelination. A series of complex intrinsic, extrinsic and epigenetic factors regulate the OPC cell cycle withdrawal, OL lineage progression and myelination. We focus on the inhibitor of DNA binding 2 (ID2), because it is widely involved in the different stages of OL differentiation and genesis. ID2 is a key transcription factor for the normal development of OL lineage cells, and the pathogenesis of WMI is closely linked with OL developmental disorders. ID4, another family member of the IDs protein, also plays a similar role in OL differentiation and genesis. ID2 and ID4 belong to the helix-loop-helix family; they lack the DNA-binding sequences and inhibit oligodendrogenesis and OPC differentiation. In this review, we mainly discuss the roles of ID2 in OL development, especially during OPC differentiation, and summarize the ID2-mediated intracellular and extracellular signaling pathways that regulate these processes. We also discuss ID4 in relation to bone morphogenetic protein signaling and oligodendrogenesis. It is likely that these developmental mechanisms are also involved in the myelin repair or remyelination in human neurological diseases.
Collapse
Affiliation(s)
- Xiaoyun Gou
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Ying Tang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Dongqiong Xiao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Junjie Ying
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| |
Collapse
|
6
|
Godoy PRDV, Montaldi APL, Sakamoto-Hojo ET. HEB silencing induces anti-proliferative effects on U87MG cells cultured as neurospheres and monolayers. Mol Med Rep 2016; 14:5253-5260. [PMID: 27779678 DOI: 10.3892/mmr.2016.5877] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 09/14/2016] [Indexed: 11/06/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a lethal tumor and novel strategies are required to overcome resistance. Transcription factor 12 (HEB) has been associated with neural and stem cell proliferation, is overexpressed in certain tumor types and is induced in irradiated U87MG cells. The present study aimed to determine whether HEB knockdown, with or without irradiation, may sensitize GBM cells. U87MG GBM and ACBRI‑371 primary human astrocytes were cultured in monolayers or neurospheres. Cell proliferation and death, cell cycle and sub‑G1 detection, and cluster of differentiation (CD) 133 immunofluorescence were analyzed by flow cytometry, whereas HEB protein expression was analyzed by immunocytochemistry and western blotting. Greater HEB protein expression was observed in U87MG neurospheres compared with ACBRI‑371, and the two cell lines exhibited nuclear HEB expression. HEB silencing in cells grown in monolayers induced a significant reduction in proliferation and decreased the proportion of cells in G0/G1 phase. In addition, HEB silencing reduced (two‑fold) the number of neurospheres compared with control scrambled (SCR) cells. HEB silencing combined with irradiation reduced U87MG cell proliferation when cultured in monolayers and reduced neurosphere cell number compared with the SCR irradiated group; however, not significantly. Differentiation of U87MG cells from neurospheres was reduced in HEB‑silenced cells, whereas in irradiated cells the proportion of CD133+ cells was similar in HEB‑silenced cells compared with the SCR control. These results suggest that HEB may contribute to the proliferation and maintenance of GBM cells. However, only limited effects were exerted by irradiation in HEB‑silenced cells. HEB may be a potential target to decrease proliferation in U87MG GBM cells, grown as monolayers or neurospheres, and may provide important information for the development of novel strategies for cancer therapy.
Collapse
Affiliation(s)
- Paulo R D V Godoy
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo 14040‑901, Brazil
| | - Ana Paula L Montaldi
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo 14040‑901, Brazil
| | - Elza T Sakamoto-Hojo
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo 14040‑901, Brazil
| |
Collapse
|
7
|
Park CK, Park I, Lee S, Sun CH, Koh Y, Park SH, Kim JE, Yun H, Lee SH. Genomic dynamics associated with malignant transformation in IDH1 mutated gliomas. Oncotarget 2016; 6:43653-66. [PMID: 26524630 PMCID: PMC4791257 DOI: 10.18632/oncotarget.6189] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 10/06/2015] [Indexed: 01/06/2023] Open
Abstract
The genomic mechanism responsible for malignant transformation remains an open question for glioma researchers, where differing conclusions have been drawn based on diverse study conditions. Therefore, it is essential to secure direct evidence using longitudinal samples from the same patient. Moreover, malignant transformation of IDH1-mutated gliomas is of potential interest, as its genomic mechanism under influence of oncometabolite remains unclear, and even higher rate of malignant transformation was reported in IDH1-mutated low grade gliomas than in wild-type IDH1 tumors. We have analyzed genomic data using next-generation sequencing technology for longitudinal samples from 3 patients with IDH1-mutated gliomas whose disease had progressed from a low grade to a high grade phenotype. Comprehensive analysis included chromosomal aberrations as well as whole exome and transcriptome sequencing, and the candidate driver genes for malignant transformation were validated with public database. Integrated analysis of genomic dynamics in clonal evolution during the malignant transformation revealed alterations in the machinery regulating gene expression, including the spliceosome complex (U2AF2), transcription factors (TCF12), and chromatin remodelers (ARID1A). Moreover, consequential expression changes implied the activation of genes associated with the restoration of the stemness of cancer cells. The alterations in genetic regulatory mechanisms may be the key factor for the major phenotypic changes in IDH1 mutated gliomas. Despite being limited to a small number of cases, this analysis provides a direct example of the genomic changes responsible for malignant transformation in gliomas.
Collapse
Affiliation(s)
- Chul-Kee Park
- Department of Neurosurgery, Seoul National University Hospital, Seoul, Korea
| | | | | | | | - Youngil Koh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - Ja Eun Kim
- Department of Neurosurgery, Seoul National University Hospital, Seoul, Korea
| | | | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
8
|
Traiffort E, Zakaria M, Laouarem Y, Ferent J. Hedgehog: A Key Signaling in the Development of the Oligodendrocyte Lineage. J Dev Biol 2016; 4:jdb4030028. [PMID: 29615592 PMCID: PMC5831774 DOI: 10.3390/jdb4030028] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 08/26/2016] [Accepted: 08/31/2016] [Indexed: 01/11/2023] Open
Abstract
The Hedgehog morphogen aroused an enormous interest since it was characterized as an essential signal for ventral patterning of the spinal cord two decades ago. The pathway is notably implicated in the initial appearance of the progenitors of oligodendrocytes (OPCs), the glial cells of the central nervous system which after maturation are responsible for axon myelination. In accordance with the requirement for Hedgehog signaling in ventral patterning, the earliest identifiable cells in the oligodendrocyte lineage are derived from the ventral ventricular zone of the developing spinal cord and brain. Here, we present the current knowledge about the involvement of Hedgehog signaling in the strict spatial and temporal regulation which characterizes the initiation and progression of the oligodendrocyte lineage. We notably describe the ability of the Hedgehog signaling to tightly orchestrate the appearance of specific combinations of genes in concert with other pathways. We document the molecular mechanisms controlling Hedgehog temporal activity during OPC specification. The contribution of the pathway to aspects of OPC development different from their specification is also highlighted especially in the optic nerve. Finally, we report the data demonstrating that Hedgehog signaling-dependency is not a universal situation for oligodendrocyte generation as evidenced in the dorsal spinal cord in contrast to the dorsal forebrain.
Collapse
Affiliation(s)
- Elisabeth Traiffort
- Neuroprotective, Neuroregenerative and Remyelinating Small Molecules' U1195, INSERM-Université Paris-Sud, Université Paris-Saclay, 80 rue du Général Leclerc, Kremlin-Bicêtre F-94276, France.
| | - Mary Zakaria
- Neuroprotective, Neuroregenerative and Remyelinating Small Molecules' U1195, INSERM-Université Paris-Sud, Université Paris-Saclay, 80 rue du Général Leclerc, Kremlin-Bicêtre F-94276, France.
| | - Yousra Laouarem
- Neuroprotective, Neuroregenerative and Remyelinating Small Molecules' U1195, INSERM-Université Paris-Sud, Université Paris-Saclay, 80 rue du Général Leclerc, Kremlin-Bicêtre F-94276, France.
| | - Julien Ferent
- IRCM, Molecular Biology of Neural Development, 110 Pine Avenue West, Montreal, QC H2W 1R7, Canada.
| |
Collapse
|
9
|
The Activators of Cyclin-Dependent Kinase 5 p35 and p39 Are Essential for Oligodendrocyte Maturation, Process Formation, and Myelination. J Neurosci 2016; 36:3024-37. [PMID: 26961956 DOI: 10.1523/jneurosci.2250-15.2016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The regulation of oligodendrocyte development and myelin formation in the CNS is poorly defined. Multiple signals influence the rate and extent of CNS myelination, including the noncanonical cyclin-dependent kinase 5 (Cdk5) whose functions are regulated by its activators p35 and p39. Here we show that selective loss of either p35 or p39 perturbed specific aspects of oligodendrocyte development, whereas loss of both p35 and p39 completely inhibited the development of mature oligodendrocytes and myelination. In the absence of p35, oligodendrocyte differentiation was delayed, process outgrowth was truncated in vitro, and the patterning and extent of myelination were perturbed in the CNS of p35(-/-) mice. In the absence of p39, oligodendrocyte maturation was transiently affected both in vitro and in vivo. However, loss of both p35 and p39 in oligodendrocyte lineage cells completely inhibited oligodendrocyte progenitor cell differentiation and myelination both in vitro and after transplantation into shiverer slice cultures. Loss of p35 and p39 had a more profound effect on oligodendrocyte development than simply the loss of Cdk5 and could not be rescued by Cdk5 overexpression. These data suggest p35 and p39 have specific and overlapping roles in oligodendrocyte development, some of which may be independent of Cdk5 activation.
Collapse
|
10
|
Tang X, Hou Y, Yang G, Wang X, Tang S, Du YE, Yang L, Yu T, Zhang H, Zhou M, Wen S, Xu L, Liu M. Stromal miR-200s contribute to breast cancer cell invasion through CAF activation and ECM remodeling. Cell Death Differ 2015; 23:132-45. [PMID: 26068592 PMCID: PMC4815985 DOI: 10.1038/cdd.2015.78] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 05/02/2015] [Accepted: 05/04/2015] [Indexed: 02/07/2023] Open
Abstract
The activation of cancer-associated fibroblasts (CAFs) is a key event in tumor progression, and alternative extracellular matrix (ECM) proteins derived from CAFs induce ECM remodeling and cancer cell invasion. Here we found that miR-200 s, which are generally downregulated in activated CAFs in breast cancer tissues and in normal fibroblasts (NFs) activated by breast cancer cells, are direct mediators of NF reprogramming into CAFs and of ECM remodeling. NFs with downregulated miR-200 s displayed the traits of activated CAFs, including accelerated migration and invasion. Ectopic expression of miR-200 s in CAFs at least partially restored the phenotypes of NFs. CAF activation may be governed by the targets of miR-200 s, Fli-1 and TCF12, which are responsible for cell development and differentiation; Fli-1 and TCF12 were obviously elevated in CAFs. Furthermore, miR-200 s and their targets influenced collagen contraction by CAFs. The upregulation of fibronectin and lysyl oxidase directly by miR-200 or indirectly through Fli-1 or TCF12 contributed to ECM remodeling, triggering the invasion and metastasis of breast cancer cells both in vitro and vivo. Thus, these data provide important and novel insights into breast CAF activation and ECM remodeling, which trigger tumor cell invasion.
Collapse
Affiliation(s)
- X Tang
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, #1 Yi-Xue-Yuan Road, Yu-zhong District, Chongqing 400016, China
| | - Y Hou
- Experimental Teaching Center of Basic Medicine Science, #1 Yi-Xue-Yuan Road, Yu-zhong District, Chongqing Medical University, Chongqing 400016, China
| | - G Yang
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 You-Yi Road, Yu-zhong District, Chongqing 400016, China
| | - X Wang
- Department of Orthopaedics, The Second Affiliated Hospital, Chongqing Medical University, No. 76 Linjiang Road, Yu-zhong District, Chongqing 400010, China
| | - S Tang
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, #1 Yi-Xue-Yuan Road, Yu-zhong District, Chongqing 400016, China
| | - Y-E Du
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, #1 Yi-Xue-Yuan Road, Yu-zhong District, Chongqing 400016, China
| | - L Yang
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, #1 Yi-Xue-Yuan Road, Yu-zhong District, Chongqing 400016, China
| | - T Yu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 You-Yi Road, Yu-zhong District, Chongqing 400016, China
| | - H Zhang
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, #1 Yi-Xue-Yuan Road, Yu-zhong District, Chongqing 400016, China
| | - M Zhou
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, #1 Yi-Xue-Yuan Road, Yu-zhong District, Chongqing 400016, China
| | - S Wen
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, #1 Yi-Xue-Yuan Road, Yu-zhong District, Chongqing 400016, China
| | - L Xu
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, #1 Yi-Xue-Yuan Road, Yu-zhong District, Chongqing 400016, China
| | - M Liu
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, #1 Yi-Xue-Yuan Road, Yu-zhong District, Chongqing 400016, China
| |
Collapse
|
11
|
Zuo H, Nishiyama A. Polydendrocytes in development and myelin repair. Neurosci Bull 2013; 29:165-76. [PMID: 23516142 DOI: 10.1007/s12264-013-1320-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Accepted: 01/30/2013] [Indexed: 11/30/2022] Open
Abstract
Polydendrocytes (NG2 cells) are a distinct type of glia that populate the developing and adult central nervous systems (CNS). In the adult CNS, they retain mitotic activity and represent the largest proliferating cell population. Genetic and epigenetic mechanisms regulate the fate of polydendrocytes, which give rise to both oligodendrocytes and astrocytes. In addition, polydendrocytes actively differentiate into myelin-forming oligodendrocytes in response to demyelination. This review summarizes the current knowledge regarding polydendrocyte development, which provides an important basis for understanding the mechanisms that lead to the remyelination of demyelinated lesions.
Collapse
Affiliation(s)
- Hao Zuo
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269-3156, USA
| | | |
Collapse
|
12
|
Lowry N, Goderie SK, Lederman P, Charniga C, Gooch MR, Gracey KD, Banerjee A, Punyani S, Silver J, Kane RS, Stern JH, Temple S. The effect of long-term release of Shh from implanted biodegradable microspheres on recovery from spinal cord injury in mice. Biomaterials 2012; 33:2892-901. [DOI: 10.1016/j.biomaterials.2011.12.048] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Accepted: 12/27/2011] [Indexed: 01/08/2023]
|
13
|
Abstract
Demyelinating disorders of the central nervous system are among the most crippling neurological diseases affecting patients at various stages of life. In the most prominent demyelinating disease, multiple sclerosis, the regeneration of myelin sheaths often fails due to a default of the resident stem/precursor cells (oligodendrocyte precursor cells) to differentiate into mature myelin forming cells. Significant advances have been made in our understanding of the molecular and cellular processes involved in remyelination. Furthermore, important insight has been gained from studies investigating the interaction of stem/precursor cells with the distinct environment of demyelinating lesions. These suggest that successful regeneration depends on a signalling environment conducive to remyelination, which is provided in the context of acute inflammation. However, multiple sclerosis lesions also contain factors that inhibit the differentiation of oligodendrocyte precursor cells into myelinating oligodendrocytes. The pattern by which remyelination inducers and inhibitors are expressed in multiple sclerosis lesions may determine a window of opportunity during which oligodendrocyte precursor cells can successfully differentiate. As the first molecules aiming at promoting remyelination are about to enter clinical trials, this review critically evaluates recent advances in our understanding of the biology of oligodendrocyte precursor cells and of the stage-dependent molecular pathology of multiple sclerosis lesions relevant to the regeneration of myelin sheaths. We propose a model that may help to provide cues for how remyelination can be therapeutically enhanced in clinical disease.
Collapse
Affiliation(s)
- Mark R Kotter
- Department of Clinical Neurosciences, MRC Centre for Stem Cells and Regenerative Medicine, University of Cambridge, Addenbrooke's Hospital, Box 167, Hills Road, Cambridge CB22QQ, UK.
| | | | | |
Collapse
|
14
|
Jang ES, Goldman JE. Pax6 expression is sufficient to induce a neurogenic fate in glial progenitors of the neonatal subventricular zone. PLoS One 2011; 6:e20894. [PMID: 21698109 PMCID: PMC3117849 DOI: 10.1371/journal.pone.0020894] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 05/11/2011] [Indexed: 11/23/2022] Open
Abstract
Background The forebrain subventricular zone (SVZ) of neonatal mammals contains a large, heterogeneous population of migratory and proliferating precursors of interneurons and glia. These cell types are produced in large numbers in the immediate postnatal period, the glioblasts populating the hemispheres with astrocytes and oligodendrocytes, the neuroblasts migrating to the olfactory bulb to become interneurons. How cell fate decisions are determined or stabilized in this mixed population is not clear, although previous studies indicate the importance of two transcription factors, Pax6 in neurons and Olig2 in glia, and suggest there may be reciprocal repression between these genes. Methodology/Principal Findings In examining the SVZ of neonatal mouse and rat brain, we find that the very large majority of SVZ cells express either Pax6 or Olig2, but few express both. We have used in vivo retro- and lenti-virus injections into the neonatal SVZ and in vitro gene transfer to demonstrate that pax6 over-expression is sufficient to down-regulate olig2 and to promote a neuronal lineage development and migration pattern in olig2-expressing cells. Furthermore, we provide evidence that Pax6 binds to the olig2 promoter and that an HEB sequence in the promoter is required for the Pax6 repression of olig2 transcription. Lastly, we constructed a lentivirus to target olig2-expressing cells in the SVZ to trace their fates, and found that the very large majority developed into glia. Conclusions/Significance We provide evidence for a direct repression of olig2 by Pax6. Since SVZ cells can display developmental plasticity in vitro, the cross-repression promotes a stabilization of cell fates. This repression may be critical in a germinal zone in which immature cells are highly migratory and are not organized into an epithelium.
Collapse
Affiliation(s)
- Eun Sook Jang
- Integrated CMBS and Pathology Graduate Programs, Columbia University, New York, New York, United States of America
| | - James E. Goldman
- Department of Pathology and Cell Biology, Columbia University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
15
|
Rivera FJ, Steffenhagen C, Kremer D, Kandasamy M, Sandner B, Couillard-Despres S, Weidner N, Küry P, Aigner L. Deciphering the oligodendrogenic program of neural progenitors: cell intrinsic and extrinsic regulators. Stem Cells Dev 2010; 19:595-606. [PMID: 19938982 DOI: 10.1089/scd.2009.0293] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In the developing and adult CNS, neural stem/progenitor cells (NSPCs) and oligodendroglial progenitor cells (OPCs) follow an oligodendrogenic process with the aim of myelinating axons. This process is to a high degree regulated by an oligodendrogenic program (OPr) composed of intrinsic and extrinsic factors that modulate the different steps required for NSPCs to differentiate into myelinating oligodendrocytes. Even though NSPCs and OPCs are present in the diseased CNS and have the capacity to generate oligodendrocytes, sparse remyelination of axons constitutes a major constraint in therapies toward multiple sclerosis (MS) and spinal cord injury (SCI). Lack of pro-oligodendrogenic factors and presence of anti-oligodendrogenic activities are thought to be the main reasons for this limitation. Thus, molecular and cellular strategies aiming at remyelination and at targeting such pro- and anti-oligodendrogenic mechanisms are currently under investigation. The present review summarizes the current knowledge on the OPr; it implements our own findings on mesenchymal stem cell-derived pro-oligodendroglial factors and on the role of p57/kip2 in oligodendroglial differentiation. Moreover, it describes molecular and cellular approaches for the development of future therapies toward remyelination.
Collapse
Affiliation(s)
- Francisco J Rivera
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
TGF-β stimulates glial-like differentiation in murine dental follicle precursor cells (mDFPCs). Neurosci Lett 2010; 471:179-84. [PMID: 20100544 DOI: 10.1016/j.neulet.2010.01.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 01/15/2010] [Accepted: 01/15/2010] [Indexed: 01/03/2023]
|
17
|
Zhang J, Li Y, Zhang ZG, Lu M, Borneman J, Buller B, Savant-Bhonsale S, Elias SB, Chopp M. Bone marrow stromal cells increase oligodendrogenesis after stroke. J Cereb Blood Flow Metab 2009; 29:1166-74. [PMID: 19384336 PMCID: PMC2849641 DOI: 10.1038/jcbfm.2009.41] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Oligodendrocytes are sensitive to ischemic damage. The Sonic hedgehog (Shh) pathway is critical in oligodendrogenesis; Gli1 is the principal effector of Shh signaling. We investigated oligodendrogenesis and Shh/Gli1 pathway activation after bone marrow stromal cell (BMSC) treatment of stroke in rats. Rats were subjected to the middle cerebral artery occlusion (MCAo). BMSCs have been shown to promote functional recovery post stroke. A therapeutic dose of BMSC (3 x 10(6) cells) treatment was initiated 1 day after MCAo. Immunohistochemistry was carried out to measure the oligodendrocyte progenitor cells, oligodendrocytes, myelin, and expressions of Shh and Gli1 at 14 days after MCAo. Gene expression of Shh and Gli1 was tested at 2 days after MCAo. An in vitro study was used to investigate the effects of BMSC on a premature oligodendrocyte cell line (N20.1 cells). BMSC treatment significantly increased O4(+) oligodendrocytes, MBP(+) area, and bromodeoxyuridine (BrdU)(+), NG2(+), BrdU(+)-NG2(+) cells, and mRNA and protein expressions of Shh and Gli1 in the ipsilateral brain of the MCAo rats than that in phosphate buffered saline (PBS)-treated rats. BMSCs promoted N20.1 cell proliferation and Gli1 mRNA expression, and these effects were abolished by the Shh pathway inhibitor cyclopamine. These data indicate that the BMSC treatment stimulates oligodendrogenesis by activation of the Shh/Gli1 pathway post stroke.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Zhang J, Chen J, Li Y, Cui X, Zheng X, Roberts C, Lu M, Elias SB, Chopp M. Niaspan treatment improves neurological functional recovery in experimental autoimmune encephalomyelitis mice. Neurobiol Dis 2008; 32:273-80. [PMID: 18778774 DOI: 10.1016/j.nbd.2008.07.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Revised: 06/25/2008] [Accepted: 07/13/2008] [Indexed: 02/02/2023] Open
Abstract
We investigated the treatment of experimental autoimmune encephalomyelitis (EAE) in mice with Niaspan, an agent used to elevate high-density lipoprotein (HDL). EAE mice were treated with Niaspan starting on the immunization or clinical onset day. Neurological functional recovery was significantly increased in the Niaspan treated mice (100 mg/kgbw) compared to the controls. Inflammatory infiltrates were significantly reduced in the Niaspan treatment group compared to the EAE controls. HDL level, intact myelin area, newly formed oligodendrocytes, regenerating axons, gene and protein levels of sonic hedgehog (Shh)/Gli1 were significantly increased in the Niaspan treated mice compared to EAE controls. These data indicate that Niaspan treatment improved functional recovery after EAE, possibly, via reducing inflammatory infiltrates and demyelination areas, and stimulating oligodendrogenesis and axonal regeneration. Niaspan-mediated activation of Shh/Gli1 pathway may promote functional recovery post-EAE.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Lowry N, Goderie SK, Adamo M, Lederman P, Charniga C, Gill J, Silver J, Temple S. Multipotent embryonic spinal cord stem cells expanded by endothelial factors and Shh/RA promote functional recovery after spinal cord injury. Exp Neurol 2008; 209:510-22. [DOI: 10.1016/j.expneurol.2007.09.031] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2007] [Accepted: 09/22/2007] [Indexed: 01/27/2023]
|
20
|
Abstract
Oligodendrocytes (OGs) assemble the myelin sheath around axons in the central nervous system. Specification of cells into the OG lineage is largely the result of interplay between bone morphogenetic protein, sonic hedgehog and Notch signaling pathways, which regulate expression of transcription factors (TFs) dictating spatial and temporal aspects of oligodendrogenesis. Many of these TFs and others then direct OG development through to a mature myelinating OG. Here we describe signaling pathways and TFs that are inductive, inhibitory, and/or permissive to OG specification and maturation. We develop a basic transcriptional network and identify similarities and differences between regulation of oligodendrogenesis in the spinal cord and brain.
Collapse
Affiliation(s)
- Danette J Nicolay
- Laboratory of Molecular Biology, College of Pharmacy and Nutrition, University of Saskatchewan, and Cameco MS Neuroscience Research Center, City Hospital, Saskatoon, Saskatchewan, Canada
| | | | | |
Collapse
|
21
|
Petryniak MA, Potter GB, Rowitch DH, Rubenstein JLR. Dlx1 and Dlx2 control neuronal versus oligodendroglial cell fate acquisition in the developing forebrain. Neuron 2007; 55:417-33. [PMID: 17678855 PMCID: PMC2039927 DOI: 10.1016/j.neuron.2007.06.036] [Citation(s) in RCA: 264] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2007] [Revised: 05/21/2007] [Accepted: 06/28/2007] [Indexed: 10/23/2022]
Abstract
Progenitors within the ventral telencephalon can generate GABAergic neurons and oligodendrocytes, but regulation of the neuron-glial switch is poorly understood. We investigated the combinatorial expression and function of Dlx1&2, Olig2, and Mash1 transcription factors in the ventral telencephalon. We show that Dlx homeobox transcription factors, required for GABAergic interneuron production, repress oligodendrocyte precursor cell (OPC) formation by acting on a common progenitor to determine neuronal versus oligodendroglial cell fate acquisition. We demonstrate that Dlx1&2 negatively regulate Olig2-dependant OPC formation and that Mash1 promotes OPC formation by restricting the number of Dlx+ progenitors. Progenitors transplanted from Dlx1&2 mutant ventral telencephalon into newborn wild-type mice do not produce neurons but differentiate into myelinating oligodendrocytes that survive into adulthood. Our results identify another role for Dlx genes as modulators of neuron versus oligodendrocyte development in the ventral embryonic forebrain.
Collapse
Affiliation(s)
- Magdalena A. Petryniak
- Nina Ireland Laboratory of Developmental Neurobiology, Department of Psychiatry, University of California at San Francisco, San Francisco, CA 94158-2611, USA
- Division of Neonatology, Department of Pediatrics, University of California at San Francisco, 533 Parnassus, San Francisco, CA, 94143-0748
| | - Gregory B. Potter
- Nina Ireland Laboratory of Developmental Neurobiology, Department of Psychiatry, University of California at San Francisco, San Francisco, CA 94158-2611, USA
- Corresponding authors: , Ph: 415-476-7872, Fax: 415-476-7884; , Ph: 415-476-7862, Fax: 415-502-7618
| | - David H. Rowitch
- Division of Neonatology, Department of Pediatrics, University of California at San Francisco, 533 Parnassus, San Francisco, CA, 94143-0748
- Institute for Regeneration Medicine, Department of Neurological Surgery, UCSF
| | - John L. R. Rubenstein
- Nina Ireland Laboratory of Developmental Neurobiology, Department of Psychiatry, University of California at San Francisco, San Francisco, CA 94158-2611, USA
- Corresponding authors: , Ph: 415-476-7872, Fax: 415-476-7884; , Ph: 415-476-7862, Fax: 415-502-7618
| |
Collapse
|
22
|
Sussman CR, Vartanian T, Miller RH. The ErbB4 neuregulin receptor mediates suppression of oligodendrocyte maturation. J Neurosci 2006; 25:5757-62. [PMID: 15958742 PMCID: PMC6724872 DOI: 10.1523/jneurosci.4748-04.2005] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Neuregulin is required for proper oligodendrocyte development, but which receptors are involved and whether neuregulin promotes or inhibits maturation remain controversial. To assess the roles of the neuregulin receptor ErbB4 in oligodendrocyte development, we examined oligodendrocyte initiation and maturation in cultures derived from erbB4 knock-out mice and rat spinal cord in the presence of neutralizing erbB4 antibodies. No differences in the development of O4+ oligodendrocytes were detected in the presence or absence of erbB4 signaling. All four epidermal growth factor receptor family members were detected in the ventral neural tube at approximately the time of initial oligodendrocyte development, consistent with redundancy in neuregulin receptor signaling at the onset of oligodendrocyte development. In contrast, greater numbers of differentiated (monoclonal antibody O1+) oligodendrocytes developed in neural tube explants from erbB4(-/-) mice than either erbB4(+/+) or erbB4(+/-) littermates as well as in cultures treated with anti-erbB4. These data indicate that ErbB4 is not required for oligodendrocyte development and, in fact, inhibits oligodendrocyte lineage maturation. Together with previous studies, these data suggest a model in which early oligodendrocyte lineage development is regulated by promiscuous neuregulin receptor signaling, but subsequent lineage progression occurs through a balance of receptor-specific promotion or inhibition of maturation.
Collapse
Affiliation(s)
- Caroline R Sussman
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio 44106-4965, USA.
| | | | | |
Collapse
|
23
|
Seifert T, Bauer J, Weissert R, Fazekas F, Storch MK. Differential expression of sonic hedgehog immunoreactivity during lesion evolution in autoimmune encephalomyelitis. J Neuropathol Exp Neurol 2005; 64:404-11. [PMID: 15892298 DOI: 10.1093/jnen/64.5.404] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The signaling molecule Sonic hedgehog (Shh) is involved in several processes of central nervous system development. Recent reports indicate that Shh expression plays a role also in certain pathologic conditions in the adult brain, including multiple sclerosis and its animal model. However, the role of Shh signaling in immune-mediated demyelinating disease remains still uncertain. The aim of our study was to investigate the distribution pattern of Shh immunoreactivity (Shh-IR) during lesion evolution in myelin-oligodendrocyte-glycoprotein-induced experimental autoimmune encephalomyelitis (MOG-EAE), a model strongly mimicking multiple sclerosis. MOG-EAE was actively induced in DA rats. Histologic evaluation was performed with light and confocal microscopy on paraffin-embedded central nervous system sections from days 20 to 120 after active immunization. Shh-IR was present within the lesions of MOG-EAE during all stages of lesion evolution. The highest staining intensity for Shh was found in remyelinating lesions. In actively demyelinating, inactive demyelinated lesions, and in remyelinating lesions, Shh-IR was detected in macrophages, endothelium, and astrocytes. Shh-IR in axons was exclusively present in remyelinating lesions. Although the exact molecular mechanisms of the Shh-signaling pathway in experimental autoimmune encephalomyelitis are yet to be determined, our findings may imply a role of Shh signaling in facilitating remyelination.
Collapse
MESH Headings
- 2',3'-Cyclic-Nucleotide Phosphodiesterases/metabolism
- Animals
- Animals, Newborn
- Brain/metabolism
- Brain/pathology
- Cell Count/methods
- Demyelinating Diseases/metabolism
- Demyelinating Diseases/pathology
- Disease Models, Animal
- Female
- Gene Expression Regulation, Developmental/physiology
- Glial Fibrillary Acidic Protein/metabolism
- Hedgehog Proteins
- Microscopy, Confocal/methods
- Myelin Proteins
- Myelin Proteolipid Protein/metabolism
- Myelin Sheath/metabolism
- Myelin Sheath/pathology
- Myelin-Associated Glycoprotein
- Myelin-Oligodendrocyte Glycoprotein
- Neuritis, Autoimmune, Experimental/chemically induced
- Neuritis, Autoimmune, Experimental/metabolism
- Neuritis, Autoimmune, Experimental/pathology
- Neurofilament Proteins/metabolism
- Neuroglia/metabolism
- Neuroglia/pathology
- Rats
- Time Factors
- Trans-Activators/metabolism
Collapse
Affiliation(s)
- Thomas Seifert
- Department of Neurology, Medical University Graz, Graz, Austria
| | | | | | | | | |
Collapse
|
24
|
Samanta J, Kessler JA. Interactions between ID and OLIG proteins mediate the inhibitory effects of BMP4 on oligodendroglial differentiation. Development 2004; 131:4131-42. [PMID: 15280210 DOI: 10.1242/dev.01273] [Citation(s) in RCA: 283] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Bone morphogenetic protein (BMP) signaling inhibits the generation of oligodendroglia and enhances generation of astrocytes by neural progenitor cells both in vitro and in vivo. This study examined the mechanisms underlying the effects of BMP signaling on glial lineage commitment. Treatment of cultured neural progenitor cells with BMP4 induced expression of all four members of the inhibitor of differentiation (ID) family of helix-loop-helix transcriptional inhibitors and blocked oligodendrocyte (OL) lineage commitment. Overexpression of Id4 or Id2 but not Id1 or Id3 in cultured progenitor cells reproduced both the inhibitory effects of BMP4 treatment on OL lineage commitment and the stimulatory effects on astrogliogenesis. Conversely, decreasing the levels of Id4 mRNA by RNA interference enhanced OL differentiation and inhibited the effects of BMP4 on glial lineage commitment. This suggests that induction of Id4 expression mediates effects of BMP signaling. Bacterial two-hybrid and co-immunoprecipitation studies demonstrated that ID4,and to a lesser extent ID2, complexed with the basic-helix-loop-helix transcription (bHLH) factors OLIG1 and OLIG2, which are required for the generation of OLs. By contrast, ID1 and ID3 did not complex with the OLIG proteins. In addition, the OLIG and ID proteins both interacted with the E2A proteins E12 and E47. Further, exposure of cultured progenitor cells to BMP4 changed the intracellular localization of OLIG1 and OLIG2 from a predominantly nuclear to a predominantly cytoplasmic localization. These observations suggest that the induction of ID4 and ID2, and their sequestration of both OLIG proteins and E2A proteins mediate the inhibitory effects of BMP signaling on OL lineage commitment and contribute to the generation of astrocytes.
Collapse
Affiliation(s)
- Jayshree Samanta
- Northwestern University's Feinberg School of Medicine, Department of Neurology, Chicago, IL 60611, USA
| | | |
Collapse
|
25
|
Affiliation(s)
- David H Rowitch
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA.
| |
Collapse
|
26
|
Miller RH, Dinsio K, Wang R, Geertman R, Maier CE, Hall AK. Patterning of spinal cord oligodendrocyte development by dorsally derived BMP4. J Neurosci Res 2004; 76:9-19. [PMID: 15048926 DOI: 10.1002/jnr.20047] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Oligodendrocyte precursors (OPCs) initially arise in the motor neuron domain of the ventral ventricular zone of the developing spinal cord. After dispersal throughout gray and white matter, OPCs differentiate in a characteristic ventral to dorsal sequence. The spatial localization of OPC induction is in part a result of both positive local sonic hedgehog signaling and dorsally derived inhibitory cues. One component of dorsal inhibitory signals seems to be members of the transforming growth factor beta (TGFbeta) superfamily such as the bone morphogenetic proteins (BMPs). We show that during the initial appearance and subsequent maturation of OPCs, BMP4 was expressed specifically in the dorsal midline and its expression was correlated spatially and temporally with phospho-Smad 1+, BMP4-responsive cells. Implantation of sonic hedgehog (Shh)-coated beads adjacent to dorsal spinal cord in Xenopus embryos induced ectopic dorsal OPCs whereas BMP4-coated beads inhibited OPC appearance. More importantly, blocking endogenous dorsal BMP4 with anti-BMP4-coated beads locally induced ectopic OPCs. Similar results were obtained using soluble ligands on slice preparations of rodent spinal cord in vitro. In dissociated cell cultures of embryonic rat spinal cord, Shh and BMP4 had antagonistic effects on OPC development and the sensitivity of oligodendrocyte lineage cells to BMP4 increased with maturation. These data suggest that BMP4 contributes to the pattern of spinal cord oligodendrogenesis by regulating both induction and maturation of spinal cord OPCs.
Collapse
Affiliation(s)
- Robert H Miller
- Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, Ohio 44106, USA.
| | | | | | | | | | | |
Collapse
|
27
|
Ohnishi A, Sawa H, Tsuda M, Sawamura Y, Itoh T, Iwasaki Y, Nagashima K. Expression of the oligodendroglial lineage-associated markers Olig1 and Olig2 in different types of human gliomas. J Neuropathol Exp Neurol 2003; 62:1052-9. [PMID: 14575240 DOI: 10.1093/jnen/62.10.1052] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Because a specific group of oligodendrogliomas is susceptible to adjuvant therapy, it is important to elucidate the biological characteristics of these tumors. In situ hybridization analyses have revealed that Olig genes are expressed in oligodendroglial lineage cells and are highly expressed in oligodendrogliomas. To clarify whether OLIG is a tumor-specific marker for oligodendrogliomas, we have investigated the expression of Olig transcripts by semiquantitative RT-PCR assay and OLIG2 protein with a new antibody in a variety of glial tumors. The semiquantitative RT-PCR revealed that high levels of expression of Olig1 and Olig2 mRNAs were present in anaplastic oligodendrogliomas and anaplastic astrocytomas, while expression of these mRNAs in grade IV glioblastomas was lower than in grade II and grade III gliomas (p < 0.01). Immunohistochemical analyses demonstrated that the mean immunopositive proportion of OLIG2 was 82% in anaplastic oligodendrogliomas but only 34% in anaplastic astrocytomas. Therefore, although OLIG2 expression was detected in a range of gliomas not specific for oligodendrogliomas, the expression level in anaplastic oligodendrogliomas was more uniform and intense than that in other glial tumors. In conclusion, combining Olig mRNA expression and immunohistochemistry of OLIG2 enables oligodendrogliomas to be distinguished from glioblastomas and other astrocytic glial tumors.
Collapse
Affiliation(s)
- Akiko Ohnishi
- Laboratory of Molecular and Cellular Pathology, Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | | | | | | | | | | | | |
Collapse
|
28
|
Dakubo GD, Wang YP, Mazerolle C, Campsall K, McMahon AP, Wallace VA. Retinal ganglion cell-derived sonic hedgehog signaling is required for optic disc and stalk neuroepithelial cell development. Development 2003; 130:2967-80. [PMID: 12756179 DOI: 10.1242/dev.00515] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The development of optic stalk neuroepithelial cells depends on Hedgehog (Hh) signaling, yet the source(s) of Hh protein in the optic stalk is unknown. We provide genetic evidence that sonic hedgehog (Shh) from retinal ganglion cells (RGCs) promotes the development of optic disc and stalk neuroepithelial cells. We demonstrate that RGCs express Shh soon after differentiation, and cells at the optic disc in close proximity to the Shh-expressing RGCs upregulate Hh target genes, which suggests they are responding to RGC-derived Shh signaling. Conditional ablation of Shh in RGCs caused a complete loss of optic disc astrocyte precursor cells, resulting in defective axon guidance in the retina, as well as conversion of the neuroepithelial cells in the optic stalk to pigmented cells. We further show that Shh signaling modulates the size of the Pax2(+) astrocyte precursor cell population at the optic disc in vitro. Together, these data provide a novel insight into the source of Hh that promotes neuroepithelial cell development in the mammalian optic disc and stalk.
Collapse
Affiliation(s)
- Gabriel D Dakubo
- Molecular Medicine Program, Ottawa Health Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
| | | | | | | | | | | |
Collapse
|