1
|
Iluta S, Nistor M, Buruiana S, Dima D. Notch and Hedgehog Signaling Unveiled: Crosstalk, Roles, and Breakthroughs in Cancer Stem Cell Research. Life (Basel) 2025; 15:228. [PMID: 40003637 PMCID: PMC11856057 DOI: 10.3390/life15020228] [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: 11/06/2024] [Revised: 01/30/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
The development of therapies that target cancer stem cells (CSCs) and bulk tumors is both crucial and urgent. Several signaling pathways, like Notch and Hedgehog (Hh), have been strongly associated with CSC stemness maintenance and metastasis. However, the extensive crosstalk present between these two signaling networks complicates the development of long-term therapies that also minimize adverse effects on healthy tissues and are not overcome by therapy resistance from CSCs. The present work aims to overview the roles of Notch and Hh in cancer outburst and the intersection of the two pathways with one another, as well as with other networks, such as Wnt/β-catenin, TGF, and JAK/STAT3, and to explore the shaping of the tumor microenvironment (TME) with specific influence on CSC development and maintenance.
Collapse
Affiliation(s)
- Sabina Iluta
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania;
| | - Madalina Nistor
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania;
| | - Sanda Buruiana
- Department of Hematology, Nicolae Testemitanu University of Medicine and Pharmacy, MD-2004 Chisinau, Moldova;
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Oncology Institute, 400015 Cluj Napoca, Romania
| |
Collapse
|
2
|
Ryu JH, Kim M, Kim A, Ro H, Kim SH, Yeo SY. Zebrafish PEX1 Is Required for the Generation of GABAergic Neuron in p3 Domain. Dev Reprod 2024; 28:129-139. [PMID: 39845513 PMCID: PMC11750165 DOI: 10.12717/dr.2024.28.4.129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/26/2024] [Accepted: 11/16/2024] [Indexed: 01/24/2025]
Abstract
Maintenance of neural progenitors requires Notch signaling in vertebrate development. Previous study has shown that Jagged2-mediated Notch signaling maintains proliferating neural progenitors in the ventral spinal cord. However, components for Jagged-mediated signaling remain poorly defined during late neurogenesis. Here we performed yeast-two hybrid screening by using the intracellular domain (ICD) of zebrafish Jagged2, and investigated a possible role of PEX1 as a component of Notch signaling for the cell-fate decision and the differentiation of neural precursors in p3 domain. Western blotting showed that zebrafish PEX1 might interacts with the ICD of zebrafish Jagged2 physically. PEX1 morpholino-injected embryos showed the increased number of GABAergic KA" neurons as well as the ectopic expression of secondary motor neurons in the p3 domain. The increased number of KA" neurons was also observed in the zebrafish embryos with PEX1 mutation induced by CRISPR/Cas9. These phenotypes resemble with that of Jagged2 morphant. Our observations imply that a critical role of PEX1 in the cell-fate decision of proliferating neural precursors in the p3 domain during the continuing growth and development of the vertebrate nervous system.
Collapse
Affiliation(s)
- Jae-Ho Ryu
- Department of Chemical and Biological
Engineering, Hanbat National University, Daejeon
34158, Korea
| | - Minjung Kim
- Department of Biological Sciences,
Sookmyung Women's University, Seoul 04310,
Korea
| | - Aseung Kim
- Department of Chemical and Biological
Engineering, Hanbat National University, Daejeon
34158, Korea
| | - Hyunju Ro
- Department of Biological Sciences,
College of Bioscience and Biotechnology, Chungnam National
University, Daejeon 34134, Korea
| | - Seok-Hyung Kim
- Sarcopenia Total Solution Center,
Wonkwang University, Iksan 54538,
Korea
| | - Sang-Yeob Yeo
- Department of Chemical and Biological
Engineering, Hanbat National University, Daejeon
34158, Korea
| |
Collapse
|
3
|
Wyart C, Carbo-Tano M, Cantaut-Belarif Y, Orts-Del'Immagine A, Böhm UL. Cerebrospinal fluid-contacting neurons: multimodal cells with diverse roles in the CNS. Nat Rev Neurosci 2023; 24:540-556. [PMID: 37558908 DOI: 10.1038/s41583-023-00723-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2023] [Indexed: 08/11/2023]
Abstract
The cerebrospinal fluid (CSF) is a complex solution that circulates around the CNS, and whose composition changes as a function of an animal's physiological state. Ciliated neurons that are bathed in the CSF - and thus referred to as CSF-contacting neurons (CSF-cNs) - are unusual polymodal interoceptive neurons. As chemoreceptors, CSF-cNs respond to variations in pH and osmolarity and to bacterial metabolites in the CSF. Their activation during infections of the CNS results in secretion of compounds to enhance host survival. As mechanosensory neurons, CSF-cNs operate together with an extracellular proteinaceous polymer known as the Reissner fibre to detect compression during spinal curvature. Once activated, CSF-cNs inhibit motor neurons, premotor excitatory neurons and command neurons to enhance movement speed and stabilize posture. At longer timescales, CSF-cNs instruct morphogenesis throughout life via the release of neuropeptides that act over long distances on skeletal muscle. Finally, recent evidence suggests that mouse CSF-cNs may act as neural stem cells in the spinal cord, inspiring new paths of investigation for repair after injury.
Collapse
Affiliation(s)
- Claire Wyart
- Institut du Cerveau (ICM), INSERM U1127, UMR CNRS 7225 Paris, Sorbonne Université, Paris, France.
| | - Martin Carbo-Tano
- Institut du Cerveau (ICM), INSERM U1127, UMR CNRS 7225 Paris, Sorbonne Université, Paris, France
| | - Yasmine Cantaut-Belarif
- Institut du Cerveau (ICM), INSERM U1127, UMR CNRS 7225 Paris, Sorbonne Université, Paris, France
| | | | - Urs L Böhm
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
4
|
Liu S, Zhang Z, Li L, Yao L, Ma Z, Li J. ADAM10- and γ-secretase-dependent cleavage of the transmembrane protein PTPRT attenuates neurodegeneration in the mouse model of Alzheimer's disease. FASEB J 2023; 37:e22734. [PMID: 36583697 DOI: 10.1096/fj.202201396r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/20/2022] [Accepted: 12/14/2022] [Indexed: 12/31/2022]
Abstract
PTPRT (receptor-type tyrosine-protein phosphatase T), a brain-specific type 1 transmembrane protein, plays an important role in neurodevelopment and synapse formation. However, whether abnormal PTPRT signaling is associated with Alzheimer's disease (AD) remains elusive. Here, we report that Ptprt mRNA expression is found to be downregulated in the brains of both human and mouse models of AD. We further identified that the PTPRT intracellular domain (PICD), which is released by ADAM10- and γ-secretase-dependent cleavage of PTPRT, efficiently translocates to the nucleus via a conserved nuclear localization signal (NLS). We show that inhibition of nuclear translocation of PICD leads to an accumulation of phosphorylated signal transducer and activator of transcription 3 (pSTAT3), a substrate of PTPRT-eventually resulting in neuronal cell death. Consistently, RNA sequencing reveals that overexpression of PICD leads to changes in the expression of genes that are functionally associated with synapse formation, cell adhesion, and protein dephosphorylation. Moreover, overexpression of PICD not only decreases the level of phospho-STAT3Y705 and amyloid β production in the hippocampus of APP/PS1 mice but also partially improves synaptic function and behavioral deficits in this mouse model of AD. These findings suggest that a novel role of the ADAM 10- and γ-secretase-dependent cleavage of PTPRT may alleviate the AD-like neurodegenerative processes.
Collapse
Affiliation(s)
- Siling Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Zhongyu Zhang
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Lianwei Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Li Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Zhanshan Ma
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jiali Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,National Institute on Drug Dependence, Peking University, Beijing, China.,IDG/McGovern Institute for Brain Research, Peking University, Beijing, China.,Kunming Primate Research Center of the Chinese Academy of Sciences, Kunming, China.,National Research Facility for Phenotypic and Genetic Analysis of Model Animals, Kunming Institute of Zoology, the Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
5
|
Vargas‐Franco D, Kalra R, Draper I, Pacak CA, Asakura A, Kang PB. The Notch signaling pathway in skeletal muscle health and disease. Muscle Nerve 2022; 66:530-544. [PMID: 35968817 PMCID: PMC9804383 DOI: 10.1002/mus.27684] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 07/20/2022] [Accepted: 07/24/2022] [Indexed: 01/05/2023]
Abstract
The Notch signaling pathway is a key regulator of skeletal muscle development and regeneration. Over the past decade, the discoveries of three new muscle disease genes have added a new dimension to the relationship between the Notch signaling pathway and skeletal muscle: MEGF10, POGLUT1, and JAG2. We review the clinical syndromes associated with pathogenic variants in each of these genes, known molecular and cellular functions of their protein products with a particular focus on the Notch signaling pathway, and potential novel therapeutic targets that may emerge from further investigations of these diseases. The phenotypes associated with two of these genes, POGLUT1 and JAG2, clearly fall within the realm of muscular dystrophy, whereas the third, MEGF10, is associated with a congenital myopathy/muscular dystrophy overlap syndrome classically known as early-onset myopathy, areflexia, respiratory distress, and dysphagia. JAG2 is a canonical Notch ligand, POGLUT1 glycosylates the extracellular domain of Notch receptors, and MEGF10 interacts with the intracellular domain of NOTCH1. Additional genes and their encoded proteins relevant to muscle function and disease with links to the Notch signaling pathway include TRIM32, ATP2A1 (SERCA1), JAG1, PAX7, and NOTCH2NLC. There is enormous potential to identify convergent mechanisms of skeletal muscle disease and new therapeutic targets through further investigations of the Notch signaling pathway in the context of skeletal muscle development, maintenance, and disease.
Collapse
Affiliation(s)
| | - Raghav Kalra
- Division of Pediatric NeurologyUniversity of Florida College of MedicineGainesvilleFlorida
| | - Isabelle Draper
- Molecular Cardiology Research InstituteTufts Medical CenterBostonMassachusetts
| | - Christina A. Pacak
- Paul and Sheila Wellstone Muscular Dystrophy CenterUniversity of Minnesota Medical SchoolMinneapolisMinnesota
- Department of NeurologyUniversity of Minnesota Medical SchoolMinneapolisMinnesota
| | - Atsushi Asakura
- Paul and Sheila Wellstone Muscular Dystrophy CenterUniversity of Minnesota Medical SchoolMinneapolisMinnesota
- Department of NeurologyUniversity of Minnesota Medical SchoolMinneapolisMinnesota
| | - Peter B. Kang
- Paul and Sheila Wellstone Muscular Dystrophy CenterUniversity of Minnesota Medical SchoolMinneapolisMinnesota
- Department of NeurologyUniversity of Minnesota Medical SchoolMinneapolisMinnesota
- Institute for Translational NeuroscienceUniversity of Minnesota Medical SchoolMinneapolisMinnesota
| |
Collapse
|
6
|
Jacobs CT, Kejriwal A, Kocha KM, Jin KY, Huang P. Temporal cell fate determination in the spinal cord is mediated by the duration of Notch signalling. Dev Biol 2022; 489:1-13. [PMID: 35623404 DOI: 10.1016/j.ydbio.2022.05.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/01/2022] [Accepted: 05/16/2022] [Indexed: 02/07/2023]
Abstract
During neural development, progenitor cells generate different types of neurons in specific time windows. Despite the characterisation of many of the transcription factor networks involved in these differentiation events, the mechanism behind their temporal regulation is poorly understood. To address this question, we studied the temporal differentiation of the simple lateral floor plate (LFP) domain in the zebrafish spinal cord. LFP progenitors generate both early-born Kolmer-Agduhr" (KA") interneuron and late-born V3 interneuron populations. Analysis using a Notch signalling reporter demonstrates that these cell populations have distinct Notch signalling profiles. Not only do V3 progenitors receive higher total levels of Notch response, but they collect this response over a longer duration compared to KA" progenitors. To test whether the duration of Notch signalling determines the temporal cell fate specification, we combined a transgene that constitutively activates Notch signalling in the ventral spinal cord with a heat shock inducible Notch signalling terminator to switch off Notch response at any given time. Sustained Notch signalling results in expanded LFP progenitors while KA" and V3 interneurons fail to specify. Early termination of Notch signalling leads to exclusively KA" cell fate, despite the high level of Notch signalling, whereas late attenuation of Notch signalling drives only V3 cell fate. This suggests that the duration of Notch signalling, not simply the level, mediates cell fate specification. Interestingly, knockdown experiments reveal a role for the Notch ligand Jag2b in maintaining LFP progenitors and limiting their differentiation into KA" and V3 interneurons. Our results indicate that Notch signalling is required for neural progenitor maintenance while a specific attenuation timetable defines the fate of the postmitotic progeny.
Collapse
Affiliation(s)
- Craig T Jacobs
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Aarti Kejriwal
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Katrinka M Kocha
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Kevin Y Jin
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Peng Huang
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
7
|
Jacobs CT, Huang P. Complex crosstalk of Notch and Hedgehog signalling during the development of the central nervous system. Cell Mol Life Sci 2021; 78:635-644. [PMID: 32880661 PMCID: PMC11072263 DOI: 10.1007/s00018-020-03627-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/23/2020] [Accepted: 08/20/2020] [Indexed: 01/20/2023]
Abstract
The development of the vertebrate central nervous system (CNS) is tightly regulated by many highly conserved cell signalling pathways. These pathways ensure that differentiation and migration events occur in a specific and spatiotemporally restricted manner. Two of these pathways, Notch and Hedgehog (Hh) signalling, have been shown to form a complex web of interaction throughout different stages of CNS development. Strikingly, some processes employ Notch signalling to regulate Hh response, while others utilise Hh signalling to modulate Notch response. Notch signalling functions upstream of Hh response through controlling the trafficking of integral pathway components as well as through modulating protein levels and transcription of downstream transcriptional factors. In contrast, Hh signalling regulates Notch response by either indirectly controlling expression of key Notch ligands and regulatory proteins or directly through transcriptional control of canonical Notch target genes. Here, we review these interactions and demonstrate the level of interconnectivity between the pathways, highlighting context-dependent modes of crosstalk. Since many other developmental signalling pathways are active in these tissues, it is likely that the interplay between Notch and Hh signalling is not only an example of signalling crosstalk but also functions as a component of a wider, multi-pathway signalling network.
Collapse
Affiliation(s)
- Craig T Jacobs
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Peng Huang
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
8
|
Yang L, Wang F, Strähle U. The Genetic Programs Specifying Kolmer-Agduhr Interneurons. Front Neurosci 2020; 14:577879. [PMID: 33162880 PMCID: PMC7581942 DOI: 10.3389/fnins.2020.577879] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/15/2020] [Indexed: 01/21/2023] Open
Abstract
Kolmer-Agduhr (KA) cells are a subgroup of interneurons positioned adjacent to the neurocoele with cilia on the apical surface protruding into the central canal of the spinal cord. Although KA cells were identified almost a century ago, their development and functions are only beginning to be unfolded. Recent studies have revealed the characteristics of KA cells in greater detail, including their spatial distribution, the timing of their differentiation, and their specification via extrinsic signaling and a unique combination of transcription factors in zebrafish and mouse. Cell lineage-tracing experiments have demonstrated that two subsets of KA cells, named KA' and KA" cells, differentiate from motoneuronal progenitors and floor-plate precursors, respectively, in both zebrafish and mouse. Although KA' and KA" cells originate from different progenitors/precursors, they each share a common set of transcription factors. Intriguingly, the combination of transcription factors that promote the acquisition of KA' cell characteristics differs from those that promote a KA" cell identity. In addition, KA' and KA" cells exhibit separable neuronal targets and differential responses to bending of the spinal cord. In this review, we summarize what is currently known about the genetic programs defining the identities of KA' and KA" cell identities. We then discuss how these two subgroups of KA cells are genetically specified.
Collapse
Affiliation(s)
- Lixin Yang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, China.,Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Feifei Wang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, China
| | - Uwe Strähle
- Institute of Biological and Chemical Systems - Biological Information Processing, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| |
Collapse
|
9
|
Chen X, Yang R, Wang J, Ruan S, Lin Z, Xin Q, Yang R, Xie J. Porcine acellular dermal matrix accelerates wound healing through miR-124-3p.1 and miR-139-5p. Cytotherapy 2020; 22:494-502. [PMID: 32571650 DOI: 10.1016/j.jcyt.2020.04.042] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/23/2020] [Accepted: 04/07/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND AIMS Cutaneous wound management is a major health problem and imposes a huge economic burden worldwide. Previous studies have demonstrated that wound healing is a highly coordinated process including epithelialization, angiogenesis, remodeling and scarring. This progression requires self-renewal, preservation and repair properties of stem cells. However, our understanding of the detailed internal regulatory mechanism following injury and the means to accelerate wound healing are limited. METHODS Our previous research revealed that porcine acellular dermal matrix (ADM) effectively promotes wound healing and scar formation through epidermal stem cells (ESCs), and this process is relevant to the alteration of internal miRNA levels. In this study, we investigated the regulatory function of porcine ADM treatment on miRNAs in ESCs. RESULTS We report that the treatment of porcine ADM reduced the levels of miR-124-3p.1 and miR-139-5p in wounds. MiR-124-3p.1 and miR-139-5p inhibited the expression of JAG1 and Notch1, respectively, by directly targeting miRNAs in ESCs. CONCLUSIONS This work demonstrates that porcine ADM induced down-regulation of miR-124-3p.1/139-5p in wounds and up-regulation of JAG1/Notch1 in ESCs, thus enhancing cutaneous wound healing.
Collapse
Affiliation(s)
- Xiaodong Chen
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan, China
| | - Ronghua Yang
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan, China
| | - Jingru Wang
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan, China
| | - Shubin Ruan
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan, China
| | - Zepeng Lin
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan, China
| | - Qi Xin
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan, China
| | - Ridong Yang
- Department of Dermatology, Guangzhou Dermatology and Prevention Institute, Guangzhou, China.
| | - Julin Xie
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
10
|
Gratton R, Tricarico PM, Moltrasio C, Lima Estevão de Oliveira AS, Brandão L, Marzano AV, Zupin L, Crovella S. Pleiotropic Role of Notch Signaling in Human Skin Diseases. Int J Mol Sci 2020; 21:E4214. [PMID: 32545758 PMCID: PMC7353046 DOI: 10.3390/ijms21124214] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023] Open
Abstract
Notch signaling orchestrates the regulation of cell proliferation, differentiation, migration and apoptosis of epidermal cells by strictly interacting with other cellular pathways. Any disruption of Notch signaling, either due to direct mutations or to an aberrant regulation of genes involved in the signaling route, might lead to both hyper- or hypo-activation of Notch signaling molecules and of target genes, ultimately inducing the onset of skin diseases. The mechanisms through which Notch contributes to the pathogenesis of skin diseases are multiple and still not fully understood. So far, Notch signaling alterations have been reported for five human skin diseases, suggesting the involvement of Notch in their pathogenesis: Hidradenitis Suppurativa, Dowling Degos Disease, Adams-Oliver Syndrome, Psoriasis and Atopic Dermatitis. In this review, we aim at describing the role of Notch signaling in the skin, particularly focusing on the principal consequences associated with its alterations in these five human skin diseases, in order to reorganize the current knowledge and to identify potential cellular mechanisms in common between these pathologies.
Collapse
Affiliation(s)
- Rossella Gratton
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (L.Z.); (S.C.)
- Department of Medical Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy
| | - Paola Maura Tricarico
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (L.Z.); (S.C.)
| | - Chiara Moltrasio
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (C.M.); (A.V.M.)
| | | | - Lucas Brandão
- Department of Pathology, Federal University of Pernambuco, Recife 50670-901, Brazil;
| | - Angelo Valerio Marzano
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (C.M.); (A.V.M.)
| | - Luisa Zupin
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (L.Z.); (S.C.)
| | - Sergio Crovella
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (L.Z.); (S.C.)
- Department of Medical Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy
| |
Collapse
|
11
|
Ringers C, Olstad EW, Jurisch-Yaksi N. The role of motile cilia in the development and physiology of the nervous system. Philos Trans R Soc Lond B Biol Sci 2019; 375:20190156. [PMID: 31884916 DOI: 10.1098/rstb.2019.0156] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Motile cilia are miniature, whip-like organelles whose beating generates a directional fluid flow. The flow generated by ciliated epithelia is a subject of great interest, as defective ciliary motility results in severe human diseases called motile ciliopathies. Despite the abundance of motile cilia in diverse organs including the nervous system, their role in organ development and homeostasis remains poorly understood. Recently, much progress has been made regarding the identity of motile ciliated cells and the role of motile-cilia-mediated flow in the development and physiology of the nervous system. In this review, we will discuss these recent advances from sensory organs, specifically the nose and the ear, to the spinal cord and brain ventricles. This article is part of the Theo Murphy meeting issue 'Unity and diversity of cilia in locomotion and transport'.
Collapse
Affiliation(s)
- Christa Ringers
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7030 Trondheim, Norway
| | - Emilie W Olstad
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7030 Trondheim, Norway
| | - Nathalie Jurisch-Yaksi
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7030 Trondheim, Norway.,Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7030 Trondheim, Norway.,Department of Neurology and Clinical Neurophysiology, St Olavs University Hospital, Edvard Griegs Gate 8, 7030 Trondheim, Norway
| |
Collapse
|
12
|
Di Bella DJ, Carcagno AL, Bartolomeu ML, Pardi MB, Löhr H, Siegel N, Hammerschmidt M, Marín-Burgin A, Lanuza GM. Ascl1 Balances Neuronal versus Ependymal Fate in the Spinal Cord Central Canal. Cell Rep 2019; 28:2264-2274.e3. [DOI: 10.1016/j.celrep.2019.07.087] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/07/2019] [Accepted: 07/23/2019] [Indexed: 01/04/2023] Open
|
13
|
Wang J, Xu SL, Duan JJ, Yi L, Guo YF, Shi Y, Li L, Yang ZY, Liao XM, Cai J, Zhang YQ, Xiao HL, Yin L, Wu H, Zhang JN, Lv SQ, Yang QK, Yang XJ, Jiang T, Zhang X, Bian XW, Yu SC. Invasion of white matter tracts by glioma stem cells is regulated by a NOTCH1-SOX2 positive-feedback loop. Nat Neurosci 2018; 22:91-105. [PMID: 30559479 DOI: 10.1038/s41593-018-0285-z] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 10/31/2018] [Indexed: 11/09/2022]
Abstract
Early invasive growth along specific anatomical structures, especially the white matter tract, is regarded as one of the main causes of poor therapeutic outcome of people with gliomas. We show that some glioma stem cells (GSCs) are preferentially located along white matter tracts, which exhibit a demyelinated phenotype, at the invasive frontier of glioma tissues. These GSCs are CD133+Notch1+, whereas the nerve fibers express the Notch ligand Jagged1. The Notch-induced transcription factor Sox9 promotes the transcription of SOX2 and the methylation level of the NOTCH1 promoter is attenuated by the upregulation of SOX2 to reinforce NOTCH1 expression in GSCs. This positive-feedback loop in a cohort of glioma subjects is correlated with a poor prognosis. Inhibition of Notch signaling attenuates the white-matter-tract tropism of GSCs. These findings provide evidence indicating that the NOTCH1-SOX2 positive-feedback loop controls GSC invasion along white matter tracts.
Collapse
Affiliation(s)
- Jun Wang
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Sen-Lin Xu
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jiang-Jie Duan
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Liang Yi
- Department of Neurosurgery, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yu-Feng Guo
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yu Shi
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lin Li
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ze-Yu Yang
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xue-Mei Liao
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jiao Cai
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yan-Qi Zhang
- Department of Medical Statistics, Military Preventive Medicine Academy, Army Medical University (Third Military Medical University), Chongqing, China
| | - Hua-Liang Xiao
- Department of Pathology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Li Yin
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Hao Wu
- Department of Radiology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jing-Na Zhang
- Department of Medical Imaging, College of Biomedical Engineering, Army Medical University (Third Military Medical University), Chongqing, China
| | - Sheng-Qing Lv
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Qing-Kai Yang
- Institute of Cancer Stem Cell, Cancer Center, Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xiao-Jun Yang
- Center for Neuroscience, Medical College, Shantou University, Shantou, China
| | - Tao Jiang
- Department of Neurosurgery, TianTan Hospital, Capital Medical University, Beijing, China
| | - Xia Zhang
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.
| | - Shi-Cang Yu
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China. .,Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.
| |
Collapse
|
14
|
Nitric oxide mediated redox regulation of protein homeostasis. Cell Signal 2018; 53:348-356. [PMID: 30408515 DOI: 10.1016/j.cellsig.2018.10.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 10/27/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022]
Abstract
Nitric oxide is a versatile diffusible signaling molecule, whose biosynthesis by three NO synthases (NOS) is tightly regulated at transcriptional and posttranslational levels, availability of co-factors, and calcium binding. Above normal levels of NO have beneficial protective effects for example in the cardiovascular system, but also contribute to the pathophysiology in the context of inflammatory diseases, and to aging and neurodegeneration in the nervous system. The effect specificity relies on the functional and spatial specificity of the NOS isoenzymes, and on the duality of two major signaling mechanisms (i) activation of soluble guanylycylase (sGC)-dependent cGMP production and (ii) direct S-nitrosylation of redox sensitive cysteines of susceptible proteins. The present review summarizes the functional implications of S-nitrosylation in the context of proteostasis, and focuses on two NO target proteins, heat shock cognate of 70 kDa (Hsc70/HSPA8) and the ubiquitin 2 ligase (UBE2D), because both are modified on functionally critical cysteines and are key regulators of chaperone mediated and assisted autophagy and proteasomal protein degradation. SNO modifications of these candidates are associated with protein accumulations and adoption of a senescent phenotype of neuronal cells suggesting that S-nitrosylations of protein homeostatic machineries contribute to aging phenomena.
Collapse
|
15
|
Hiscock TW, Miesfeld JB, Mosaliganti KR, Link BA, Megason SG. Feedback between tissue packing and neurogenesis in the zebrafish neural tube. Development 2018; 145:dev.157040. [PMID: 29678815 DOI: 10.1242/dev.157040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 04/03/2018] [Indexed: 01/22/2023]
Abstract
Balancing the rate of differentiation and proliferation in developing tissues is essential to produce organs of robust size and composition. Although many molecular regulators have been established, how these connect to physical and geometrical aspects of tissue architecture is poorly understood. Here, using high-resolution timelapse imaging, we find that changes to cell geometry associated with dense tissue packing play a significant role in regulating differentiation rate in the zebrafish neural tube. Specifically, progenitors that are displaced away from the apical surface due to crowding, tend to differentiate in a Notch-dependent manner. Using simulations we show that interplay between progenitor density, cell shape and changes in differentiation rate could naturally result in negative-feedback control on progenitor cell number. Given these results, we suggest a model whereby differentiation rate is regulated by density dependent effects on cell geometry to: (1) correct variability in cell number; and (2) balance the rates of proliferation and differentiation over development to 'fill' the available space.
Collapse
Affiliation(s)
- Tom W Hiscock
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Joel B Miesfeld
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | - Brian A Link
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Sean G Megason
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
16
|
Chen XD, Ruan SB, Lin ZP, Zhou Z, Zhang FG, Yang RH, Xie JL. Effects of porcine acellular dermal matrix treatment on wound healing and scar formation: Role of Jag1 expression in epidermal stem cells. Organogenesis 2018; 14:25-35. [PMID: 29420128 DOI: 10.1080/15476278.2018.1436023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Skin wound healing involves Notch/Jagged1 signaling. However, little is known how Jag1 expression level in epidermal stem cells (ESCs) contributes to wound healing and scar formation. We applied multiple cellular and molecular techniques to examine how Jag1 expression in ESCs modulates ESCs differentiation to myofibroblasts (MFB) in vitro, interpret how Jag1 expression in ESCs is involved in wound healing and scar formation in mice, and evaluate the effects of porcine acellular dermal matrix (ADM) treatment on wound healing and scar formation. We found that Jag1, Notch1 and Hes1 expression was up-regulated in the wound tissue during the period of wound healing. Furthermore, Jag1 expression level in the ESCs was positively associated with the level of differentiation to MFB. ESC-specific knockout of Jag1 delayed wound healing and promoted scar formation in vivo. In addition, we reported that porcine ADM treatment after skin incision could accelerate wound closure and reduce scar formation in vivo. This effect was associated with decreased expression of MFB markers, including α-SMA Col-1 and Col-III in wound tissues. Finally, we confirmed that porcine ADM treatment could increase Jag1, Notch1 and Hesl expression in wound tissues. Taken together, our results suggested that ESC-specific Jag1 expression levels are critical for wound healing and scar formation, and porcine ADM treatment would be beneficial in promoting wound healing and preventing scar formation by enhancing Notch/Jagged1 signaling pathway in ESCs.
Collapse
Affiliation(s)
- Xiao-Dong Chen
- a Department of Burn Surgery , The First People's Hospital of Foshan , Foshan , Guangdong , China
| | - Shu-Bin Ruan
- a Department of Burn Surgery , The First People's Hospital of Foshan , Foshan , Guangdong , China
| | - Ze-Peng Lin
- a Department of Burn Surgery , The First People's Hospital of Foshan , Foshan , Guangdong , China
| | - Ziheng Zhou
- b Department of Burn Surgery , First Affiliated Hospital of Sun Yat-Sen University , Guangzhou , Guangdong , China
| | - Feng-Gang Zhang
- a Department of Burn Surgery , The First People's Hospital of Foshan , Foshan , Guangdong , China
| | - Rong-Hua Yang
- a Department of Burn Surgery , The First People's Hospital of Foshan , Foshan , Guangdong , China
| | - Ju-Lin Xie
- b Department of Burn Surgery , First Affiliated Hospital of Sun Yat-Sen University , Guangzhou , Guangdong , China
| |
Collapse
|
17
|
Kong HJ, Ryu JH, Kim J, Kim JW, Seong B, Whang I, Park JY, Yeo SY. Generation of motor neurons requires spatiotemporal coordination between retinoic acid and Mib-mediated Notch signaling. Anim Cells Syst (Seoul) 2018; 22:76-81. [PMID: 30460083 PMCID: PMC6138348 DOI: 10.1080/19768354.2018.1443494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 01/18/2018] [Accepted: 02/13/2018] [Indexed: 11/08/2022] Open
Abstract
Mind bomb (Mib) is an E3 ubiquitin ligase that activates the Notch signaling pathway. A previous study demonstrated that the generation of late-born GABAergic neurons may be regulated by the interplay between Mib and retinoic acid (RA). However, the relationship between Mib function and the retinoid pathway during the generation of late-born motor neurons remains unclear. We investigated the differentiation of neural progenitors into motor neurons by inhibition of Notch signaling and administration of RA to Tg[hsp70-Mib:EGFP] embryos. The number of motor neurons in the ventral spinal cord increased or decreased depending on the temporal inhibition of Mib-mediated Notch signaling. Inhibition of the retinoid pathway by citral treatment had a synergistic effect with overexpression of Mib:EGFP on the generation of ectopic motor neurons. Additionally, the proteolytic fragment of Mib was detected in differentiated P19 cells following treatment with RA. Our observations imply that the function of Mib may be attenuated by the retinoid pathway, and that Mib-mediated Notch signaling and the retinoid pathway play critical roles in the spatiotemporal differentiation of motor neurons.
Collapse
Affiliation(s)
- Hee Jeong Kong
- Biotechnology Research Division, National Institute of Fisheries Science, Busan, Korea
| | - Jae-Ho Ryu
- Department of Chemical and Biological Engineering, Hanbat National University, Daejeon, Korea
| | - Julan Kim
- Biotechnology Research Division, National Institute of Fisheries Science, Busan, Korea
| | - Ju-Won Kim
- Biotechnology Research Division, National Institute of Fisheries Science, Busan, Korea
| | - Bomi Seong
- Department of Chemical and Biological Engineering, Hanbat National University, Daejeon, Korea
| | - Ilson Whang
- Department of Genetic Resources Research, National Marine Biodiversity Institute of Korea (MABIK), Seochun, Korea
| | - Jung Youn Park
- Biotechnology Research Division, National Institute of Fisheries Science, Busan, Korea
| | - Sang-Yeob Yeo
- Department of Chemical and Biological Engineering, Hanbat National University, Daejeon, Korea
| |
Collapse
|
18
|
Cardozo MJ, Mysiak KS, Becker T, Becker CG. Reduce, reuse, recycle – Developmental signals in spinal cord regeneration. Dev Biol 2017; 432:53-62. [DOI: 10.1016/j.ydbio.2017.05.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 02/03/2017] [Accepted: 05/11/2017] [Indexed: 02/06/2023]
|
19
|
Djenoune L, Wyart C. Light on a sensory interface linking the cerebrospinal fluid to motor circuits in vertebrates. J Neurogenet 2017; 31:113-127. [PMID: 28789587 DOI: 10.1080/01677063.2017.1359833] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The cerebrospinal fluid (CSF) is circulating around the entire central nervous system (CNS). The main function of the CSF has been thought to insure the global homeostasis of the CNS. Recent evidence indicates that the CSF also dynamically conveys signals modulating the development and the activity of the nervous system. The later observation implies that cues from the CSF could act on neurons in the brain and the spinal cord via bordering receptor cells. Candidate neurons to enable such modulation are the cerebrospinal fluid-contacting neurons (CSF-cNs) that are located precisely at the interface between the CSF and neuronal circuits. The atypical apical extension of CSF-cNs bears a cluster of microvilli bathing in the CSF indicating putative sensory or secretory roles in relation with the CSF. In the brainstem and spinal cord, CSF-cNs have been described in over two hundred species by Kolmer and Agduhr, suggesting an important function within the spinal cord. However, the lack of specific markers and the difficulty to access CSF-cNs hampered their physiological investigation. The transient receptor potential channel PKD2L1 is a specific marker of spinal CSF-cNs in vertebrate species. The transparency of zebrafish at early stages eases the functional characterization of pkd2l1+ CSF-cNs. Recent studies demonstrate that spinal CSF-cNs detect spinal curvature via the channel PKD2L1 and modulate locomotion and posture by projecting onto spinal interneurons and motor neurons in vivo. In vitro recordings demonstrated that spinal CSF-cNs are sensing pH variations mainly through ASIC channels, in combination with PKD2L1. Altogether, neurons contacting the CSF appear as a novel sensory modality enabling the detection of mechanical and chemical stimuli from the CSF and modulating the excitability of spinal circuits underlying locomotion and posture.
Collapse
Affiliation(s)
- Lydia Djenoune
- a Institut du Cerveau et de la Moelle épinière (ICM) , Paris , France
| | - Claire Wyart
- a Institut du Cerveau et de la Moelle épinière (ICM) , Paris , France
| |
Collapse
|
20
|
Wang P, Shu B, Xu Y, Zhu J, Liu J, Zhou Z, Chen L, Zhao J, Liu X, Qi S, Xiong K, Xie J. Basic fibroblast growth factor reduces scar by inhibiting the differentiation of epidermal stem cells to myofibroblasts via the Notch1/Jagged1 pathway. Stem Cell Res Ther 2017; 8:114. [PMID: 28511663 PMCID: PMC5434520 DOI: 10.1186/s13287-017-0549-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/28/2017] [Accepted: 03/31/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Basic fibroblast growth factor (bFGF) plays an important role in promoting wound healing and reducing scar, but the possible molecular mechanisms are still unclear. Our previous studies have found that activating the Notch1/Jagged1 pathway can inhibit the differentiation of epidermal stem cells (ESCs) to myofibroblasts (MFB). Herein, we document that bFGF reduces scar by inhibiting the differentiation of ESCs to MFB via activating the Notch1/Jagged1 pathway. METHODS In in-vitro study, ESCs were isolated from 10 neonatal SD rats (1-3 days old), cultured in keratinocyte serum-free medium, and divided into six groups: bFGF group, bFGF + SU5402 group, bFGF + DAPT group, siJagged1 group, bFGF + siJagged1 group, and control group. Jagged1 of the ESCs in the siJagged1 group and bFGF + siJagged1 group was knocked down by small-interfering RNA transfection. Expression of ESC markers (CK15/CK10), MFB markers (α-SMA, Collagen I, Collagen III), and Notch1/Jagged1 components (Jagged1, Notch1, Hes1) was detected by FCM, qRT-PCR, and western blot analysis to study the relationships of bFGF, ESCs, and Notch1/Jagged1 pathway. In in-vivo study, the wound healing time and scar hyperplasia were observed on rabbit ear scar models. The quality of wound healing was estimated by hematoxylin and eosin staining and Masson staining. Expression of ESC markers, MFB markers and Notch1/Jagged1 components was elucidated by immunohistochemistry, immunofluorescence, and western blot analysis. RESULTS The in-vitro study showed that bFGF could significantly upregulate the expression of ESC markers and Notch1/Jagged1 components, while downregulating the expression of MFB markers at the same time. However, these effects could be obviously decreased when we knocked down Jagged1 or added DAPT. Similarly, in in-vivo study, bFGF also exhibited its functions in inhibiting the differentiation of rabbit ESCs to MFB by activating the Notch1/Jagged1 pathway, which improved the wound healing quality and alleviated scar significantly. CONCLUSION These results provide evidence that bFGF can reduce scar by inhibiting the differentiation of ESCs to MFB via the Notch1/Jagged1 pathway, and present a new promising potential direction for the treatment of scar.
Collapse
Affiliation(s)
- Peng Wang
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| | - Bin Shu
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| | - Yingbin Xu
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| | - Jiayuan Zhu
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| | - Jian Liu
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| | - Ziheng Zhou
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| | - Lei Chen
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| | - Jingling Zhao
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| | - Xusheng Liu
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| | - Shaohai Qi
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan Province 410013 People’s Republic of China
| | - Julin Xie
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| |
Collapse
|
21
|
Petracca YL, Sartoretti MM, Di Bella DJ, Marin-Burgin A, Carcagno AL, Schinder AF, Lanuza GM. The late and dual origin of cerebrospinal fluid-contacting neurons in the mouse spinal cord. Development 2016; 143:880-91. [PMID: 26839365 DOI: 10.1242/dev.129254] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 01/25/2016] [Indexed: 12/16/2022]
Abstract
Considerable progress has been made in understanding the mechanisms that control the production of specialized neuronal types. However, how the timing of differentiation contributes to neuronal diversity in the developing spinal cord is still a pending question. In this study, we show that cerebrospinal fluid-contacting neurons (CSF-cNs), an anatomically discrete cell type of the ependymal area, originate from surprisingly late neurogenic events in the ventral spinal cord. CSF-cNs are identified by the expression of the transcription factors Gata2 and Gata3, and the ionic channels Pkd2l1 and Pkd1l2. Contrasting with Gata2/3(+) V2b interneurons, differentiation of CSF-cNs is independent of Foxn4 and takes place during advanced developmental stages previously assumed to be exclusively gliogenic. CSF-cNs are produced from two distinct dorsoventral regions of the mouse spinal cord. Most CSF-cNs derive from progenitors circumscribed to the late-p2 and the oligodendrogenic (pOL) domains, whereas a second subset of CSF-cNs arises from cells bordering the floor plate. The development of these two subgroups of CSF-cNs is differentially controlled by Pax6, they adopt separate locations around the postnatal central canal and they display electrophysiological differences. Our results highlight that spatiotemporal mechanisms are instrumental in creating neural cell diversity in the ventral spinal cord to produce distinct classes of interneurons, motoneurons, CSF-cNs, glial cells and ependymal cells.
Collapse
Affiliation(s)
- Yanina L Petracca
- Developmental Neurobiology Lab, Instituto Leloir and Consejo Nacional de Investigaciones Científicas y Técnicas (IIBBA-CONICET), Avenida Patricias Argentinas 435, Buenos Aires 1405, Argentina
| | - Maria Micaela Sartoretti
- Developmental Neurobiology Lab, Instituto Leloir and Consejo Nacional de Investigaciones Científicas y Técnicas (IIBBA-CONICET), Avenida Patricias Argentinas 435, Buenos Aires 1405, Argentina
| | - Daniela J Di Bella
- Developmental Neurobiology Lab, Instituto Leloir and Consejo Nacional de Investigaciones Científicas y Técnicas (IIBBA-CONICET), Avenida Patricias Argentinas 435, Buenos Aires 1405, Argentina
| | - Antonia Marin-Burgin
- Neuronal Plasticity Lab, Instituto Leloir and Consejo Nacional de Investigaciones Científicas y Técnicas (IIBBA-CONICET), Avenida Patricias Argentinas 435, Buenos Aires 1405, Argentina
| | - Abel L Carcagno
- Developmental Neurobiology Lab, Instituto Leloir and Consejo Nacional de Investigaciones Científicas y Técnicas (IIBBA-CONICET), Avenida Patricias Argentinas 435, Buenos Aires 1405, Argentina
| | - Alejandro F Schinder
- Neuronal Plasticity Lab, Instituto Leloir and Consejo Nacional de Investigaciones Científicas y Técnicas (IIBBA-CONICET), Avenida Patricias Argentinas 435, Buenos Aires 1405, Argentina
| | - Guillermo M Lanuza
- Developmental Neurobiology Lab, Instituto Leloir and Consejo Nacional de Investigaciones Científicas y Técnicas (IIBBA-CONICET), Avenida Patricias Argentinas 435, Buenos Aires 1405, Argentina
| |
Collapse
|
22
|
Ryu JH, Kong HJ, Park JY, Lim KE, An CM, Lee J, Yeo SY. Generation of late-born neurons in the ventral spinal cord requires the coordination of retinoic acid and Notch signaling. Neurosci Lett 2015; 602:95-8. [PMID: 26151587 DOI: 10.1016/j.neulet.2015.06.051] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 06/26/2015] [Accepted: 06/29/2015] [Indexed: 11/25/2022]
Abstract
Neural progenitor cells generate various types of neurons and glia in a tightly regulated manner. During primary neurogenesis, retinoic acid (RA) acts earlier than Notch signaling and regulates differentiation and proliferation by upregulating proneural and neurogenic genes in the neural plate. However, the relationship between Notch signaling and the retinoid pathway during late neurogenesis remains unclear. We investigated the role of Mindbomb (Mib)-mediated Notch signaling in the differentiation of neural progenitors during late neurogenesis by overexpressing Mib and administering RA to Tg[hsp70-Mib:EGFP]. The majority of cells in the p3 domain differentiated into GABAergic Kolmer-Agduhr (KA) cells in Tg[hsp70-mib:EGFP] embryos heat-shocked during late neurogenesis, whereas these phenotypes were suppressed by exogenous RA. Our observations suggest that Mib-mediated Notch signaling plays a critical role in the temporal differentiation of neural progenitors, and that the generation of late-born KA″ cells is regulated by the interplay between Mib and RA.
Collapse
Affiliation(s)
- Jae-Ho Ryu
- Department of Chemical and Biological Engineering, Hanbat National University, Daejeon 305-719, South Korea; Genomic Design Bioengineering Company, Daejeon 306-230, South Korea
| | - Hee Jeong Kong
- Biotechnology Research Division, National Fisheries Research and Development Institute, Busan 619-705, South Korea
| | - Jung Youn Park
- Biotechnology Research Division, National Fisheries Research and Development Institute, Busan 619-705, South Korea
| | - Kyung-Eun Lim
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cheul Min An
- Biotechnology Research Division, National Fisheries Research and Development Institute, Busan 619-705, South Korea
| | - Jehee Lee
- Department of Marine Life Sciences, School of Marine Biomedical Science, Jeju National University, Jeju 690-756, South Korea
| | - Sang-Yeob Yeo
- Department of Chemical and Biological Engineering, Hanbat National University, Daejeon 305-719, South Korea.
| |
Collapse
|
23
|
Djenoune L, Khabou H, Joubert F, Quan FB, Nunes Figueiredo S, Bodineau L, Del Bene F, Burcklé C, Tostivint H, Wyart C. Investigation of spinal cerebrospinal fluid-contacting neurons expressing PKD2L1: evidence for a conserved system from fish to primates. Front Neuroanat 2014; 8:26. [PMID: 24834029 PMCID: PMC4018565 DOI: 10.3389/fnana.2014.00026] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 04/10/2014] [Indexed: 12/11/2022] Open
Abstract
Over 90 years ago, Kolmer and Agduhr identified spinal cerebrospinal fluid-contacting neurons (CSF-cNs) based on their morphology and location within the spinal cord. In more than 200 vertebrate species, they observed ciliated neurons around the central canal that extended a brush of microvilli into the cerebrospinal fluid (CSF). Although their morphology is suggestive of a primitive sensory cell, their function within the vertebrate spinal cord remains unknown. The identification of specific molecular markers for these neurons in vertebrates would benefit the investigation of their physiological roles. PKD2L1, a transient receptor potential channel that could play a role as a sensory receptor, has been found in cells contacting the central canal in mouse. In this study, we demonstrate that PKD2L1 is a specific marker for CSF-cNs in the spinal cord of mouse (Mus musculus), macaque (Macaca fascicularis) and zebrafish (Danio rerio). In these species, the somata of spinal PKD2L1+ CSF-cNs were located below or within the ependymal layer and extended an apical bulbous extension into the central canal. We found GABAergic PKD2L1-expressing CSF-cNs in all three species. We took advantage of the zebrafish embryo for its transparency and rapid development to identify the progenitor domains from which pkd2l1+ CSF-cNs originate. pkd2l1+ CSF-cNs were all GABAergic and organized in two rows—one ventral and one dorsal to the central canal. Their location and marker expression is consistent with previously described Kolmer–Agduhr cells. Accordingly, pkd2l1+ CSF-cNs were derived from the progenitor domains p3 and pMN defined by the expression of nkx2.2a and olig2 transcription factors, respectively. Altogether our results suggest that a system of CSF-cNs expressing the PKD2L1 channel is conserved in the spinal cord across bony vertebrate species.
Collapse
Affiliation(s)
- Lydia Djenoune
- Institut du Cerveau et de la Moelle Épinière, Hôpital de la Pitié-Salpêtrière Paris, France ; Institut National de la Santé et de la Recherche Médicale UMR 1127 Paris, France ; Centre National de la Recherche Scientifique UMR 7225 Paris, France ; UPMC Univ. Paris 06 Paris, France ; Muséum National d'Histoire Naturelle Paris, France ; Centre National de la Recherche Scientifique UMR 7221 Paris, France
| | - Hanen Khabou
- Institut du Cerveau et de la Moelle Épinière, Hôpital de la Pitié-Salpêtrière Paris, France ; Institut National de la Santé et de la Recherche Médicale UMR 1127 Paris, France ; Centre National de la Recherche Scientifique UMR 7225 Paris, France ; UPMC Univ. Paris 06 Paris, France
| | - Fanny Joubert
- UPMC Univ. Paris 06 Paris, France ; Institut National de la Santé et de la Recherche Médicale UMR S 1158 Paris, France
| | - Feng B Quan
- Muséum National d'Histoire Naturelle Paris, France ; Centre National de la Recherche Scientifique UMR 7221 Paris, France
| | - Sophie Nunes Figueiredo
- Institut du Cerveau et de la Moelle Épinière, Hôpital de la Pitié-Salpêtrière Paris, France ; Institut National de la Santé et de la Recherche Médicale UMR 1127 Paris, France ; Centre National de la Recherche Scientifique UMR 7225 Paris, France ; UPMC Univ. Paris 06 Paris, France
| | - Laurence Bodineau
- UPMC Univ. Paris 06 Paris, France ; Institut National de la Santé et de la Recherche Médicale UMR S 1158 Paris, France
| | - Filippo Del Bene
- Institut Curie Paris, France ; Centre National de la Recherche Scientifique UMR 3215 Paris, France ; Institut National de la Santé et de la Recherche Médicale U 934 Paris, France
| | - Céline Burcklé
- Institut du Cerveau et de la Moelle Épinière, Hôpital de la Pitié-Salpêtrière Paris, France ; Institut National de la Santé et de la Recherche Médicale UMR 1127 Paris, France ; Centre National de la Recherche Scientifique UMR 7225 Paris, France ; UPMC Univ. Paris 06 Paris, France
| | - Hervé Tostivint
- Muséum National d'Histoire Naturelle Paris, France ; Centre National de la Recherche Scientifique UMR 7221 Paris, France
| | - Claire Wyart
- Institut du Cerveau et de la Moelle Épinière, Hôpital de la Pitié-Salpêtrière Paris, France ; Institut National de la Santé et de la Recherche Médicale UMR 1127 Paris, France ; Centre National de la Recherche Scientifique UMR 7225 Paris, France ; UPMC Univ. Paris 06 Paris, France
| |
Collapse
|
24
|
Chen G, Qiu Y, Sun L, Yu M, Wang W, Xiao W, Yang Y, Liu Y, Yang S, Teitelbaum DH, Ma Y, Lu D, Yang H. The jagged-2/notch-1/hes-1 pathway is involved in intestinal epithelium regeneration after intestinal ischemia-reperfusion injury. PLoS One 2013; 8:e76274. [PMID: 24098462 PMCID: PMC3789708 DOI: 10.1371/journal.pone.0076274] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 08/26/2013] [Indexed: 12/27/2022] Open
Abstract
Background Notch signaling plays a critical role in the maintenance of intestinal crypt epithelial cell proliferation. The aim of this study was to investigate the role of Notch signaling in the proliferation and regeneration of intestinal epithelium after intestinal ischemia reperfusion (I/R) injury. Methods Male Sprague-Dawley rats were subjected to sham operation or I/R by occlusion of the superior mesenteric artery (SMA) for 20 min. Intestinal tissue samples were collected at 0, 1, 2, 4, and 6 h after reperfusion. Proliferation of the intestinal epithelium was evaluated by immunohistochemical staining of proliferating nuclear antigen (PCNA). The mRNA and protein expression levels of Notch signaling components were examined using Real-time PCR and Western blot analyses. Immunofluorescence was also performed to detect the expression and location of Jagged-2, cleaved Notch-1, and Hes-1 in the intestine. Finally, the γ-secretase inhibitor DAPT and the siRNA for Jagged-2 and Hes-1 were applied to investigate the functional role of Notch signaling in the proliferation of intestinal epithelial cells in an in vitro IEC-6 culture system. Results I/R injury caused increased intestinal crypt epithelial cell proliferation and increased mRNA and protein expression of Jagged-2, Notch-1, and Hes-1. The immunofluorescence results further confirmed increased protein expression of Jagged-2, cleaved Notch-1, and Hes-1 in the intestinal crypts. The inhibition of Notch signaling with DAPT and the suppression of Jagged-2 and Hes-1 expression using siRNA both significantly inhibited the proliferation of IEC-6 cells. Conclusion The Jagged-2/Notch-1/Hes-1 signaling pathway is involved in intestinal epithelium regeneration early after I/R injury by increasing crypt epithelial cell proliferation.
Collapse
Affiliation(s)
- Guoqing Chen
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yuan Qiu
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Lihua Sun
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Min Yu
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Wensheng Wang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yang Yang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yong Liu
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Songwei Yang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Daniel H. Teitelbaum
- Department of Surgery, the University of Michigan Medical School, Ann Arbor, Michigan
| | - Yuanhang Ma
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Dingsong Lu
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- * E-mail:
| |
Collapse
|
25
|
Chakravarty S, Reddy BR, Sudhakar SR, Saxena S, Das T, Meghah V, Brahmendra Swamy CV, Kumar A, Idris MM. Chronic unpredictable stress (CUS)-induced anxiety and related mood disorders in a zebrafish model: altered brain proteome profile implicates mitochondrial dysfunction. PLoS One 2013; 8:e63302. [PMID: 23691016 PMCID: PMC3653931 DOI: 10.1371/journal.pone.0063302] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 04/02/2013] [Indexed: 11/18/2022] Open
Abstract
Anxiety and depression are major chronic mood disorders, and the etiopathology for each appears to be repeated exposure to diverse unpredictable stress factors. Most of the studies on anxiety and related mood disorders are performed in rodents, and a good model is chronic unpredictable stress (CUS). In this study, we have attempted to understand the molecular basis of the neuroglial and behavioral changes underlying CUS-induced mood disorders in the simplest vertebrate model, the zebrafish, Danio rerio. Zebrafish were subjected to a CUS paradigm in which two different stressors were used daily for 15 days, and thorough behavioral analyses were performed to assess anxiety and related mood disorder phenotypes using the novel tank test, shoal cohesion and scototaxis. Fifteen days of exposure to chronic stressors appears to induce an anxiety and related mood disorder phenotype. Decreased neurogenesis, another hallmark of anxiety and related disorders in rodents, was also observed in this zebrafish model. The common molecular markers of rodent anxiety and related disorders, corticotropin-releasing factor (CRF), calcineurin (ppp3r1a) and phospho cyclic AMP response element binding protein (pCREB), were also replicated in the fish model. Finally, using 2DE FTMS/ITMSMS proteomics analyses, 18 proteins were found to be deregulated in zebrafish anxiety and related disorders. The most affected process was mitochondrial function, 4 of the 18 differentially regulated proteins were mitochondrial proteins: PHB2, SLC25A5, VDAC3 and IDH2, as reported in rodent and clinical samples. Thus, the zebrafish CUS model and proteomics can facilitate not only uncovering new molecular targets of anxiety and related mood disorders but also the routine screening of compounds for drug development.
Collapse
Affiliation(s)
- Sumana Chakravarty
- Chemical Biology Division, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad, India
- * E-mail: (SC); (MMI)
| | - Bommana R. Reddy
- Chemical Biology Division, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad, India
| | - Sreesha R. Sudhakar
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Habsiguda, Hyderabad, India
| | - Sandeep Saxena
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Habsiguda, Hyderabad, India
| | - Tapatee Das
- Chemical Biology Division, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad, India
| | - Vuppalapaty Meghah
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Habsiguda, Hyderabad, India
| | | | - Arvind Kumar
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Habsiguda, Hyderabad, India
| | - Mohammed M. Idris
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Habsiguda, Hyderabad, India
- * E-mail: (SC); (MMI)
| |
Collapse
|
26
|
Kang K, Lee D, Hong S, Park SG, Song MR. The E3 ligase Mind bomb-1 (Mib1) modulates Delta-Notch signaling to control neurogenesis and gliogenesis in the developing spinal cord. J Biol Chem 2012; 288:2580-92. [PMID: 23223237 DOI: 10.1074/jbc.m112.398263] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The Notch signaling pathway is essential for neuronal and glial specification during CNS development. Mind bomb-1 (Mib1) is an E3 ubiquitin ligase that ubiquitinates and promotes the endocytosis of Notch ligands. Although Mib1 is essential for transmitting the Notch signal, it is still unclear whether it is a primary regulator of Notch ligand activity in the developing spinal cord. In Mib1 conditional knock-out mice, we observed depletion of spinal progenitors, premature differentiation of neurons, and unbalanced specification of V2 interneurons, all of which mimic the conventional Notch phenotype. In agreement with this, the reduction of progenitors in the absence of Mib1 led to a loss of both astrocytes and oligodendrocytes. Late removal of Mib1 using a drug-inducible system suppressed glial differentiation, suggesting that Mib1 continues to play a role in the formation of late progenitors mainly designated for gliogenesis. Finally, misexpression of Mib1 or Mib1 deletion mutants revealed that the ring domain of Mib1 is required for the specification of V2 interneurons in the chick neural tube. Together, these findings suggest that Mib1 is a major component of the signal-sending cells required to provide Notch ligand activity for specifying neurons and glia in the spinal cord.
Collapse
Affiliation(s)
- Kyungjoon Kang
- School of Life Sciences, Bioimaging Research Center, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju 500-712, Republic of Korea
| | | | | | | | | |
Collapse
|
27
|
Zannino DA, Sagerström CG, Appel B. olig2-Expressing hindbrain cells are required for migrating facial motor neurons. Dev Dyn 2012; 241:315-26. [PMID: 22275004 DOI: 10.1002/dvdy.23718] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The complicated trajectory of facial motor neuron migration requires coordination of intrinsic signals and cues from the surrounding environment. Migration begins in rhombomere (r) 4 where facial motor neurons are born and proceeds in a caudal direction. Once facial motor neurons reach their target rhombomeres, they migrate laterally and radially from the ventral neural tube. In zebrafish, as facial motor neurons migrate through r5/r6, they pass near cells that express olig2, which encodes a bHLH transcription factor. In this study, we found that olig2 function is required for facial motor neurons to complete their caudal migration into r6 and r7 and form stereotypical clusters. Additionally, embryos that lack mafba function, in which facial motor neurons also fail to complete caudal migration, lack olig2 expression in r5 and r6. Our data raise the possibility that cells expressing olig2 are intermediate targets that help guide facial motor neuron migration.
Collapse
Affiliation(s)
- Denise A Zannino
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | | | | |
Collapse
|
28
|
Attenuation of Notch and Hedgehog signaling is required for fate specification in the spinal cord. PLoS Genet 2012; 8:e1002762. [PMID: 22685423 PMCID: PMC3369957 DOI: 10.1371/journal.pgen.1002762] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2011] [Accepted: 04/30/2012] [Indexed: 02/01/2023] Open
Abstract
During the development of the spinal cord, proliferative neural progenitors differentiate into postmitotic neurons with distinct fates. How cells switch from progenitor states to differentiated fates is poorly understood. To address this question, we studied the differentiation of progenitors in the zebrafish spinal cord, focusing on the differentiation of Kolmer-Agduhr″ (KA″) interneurons from lateral floor plate (LFP) progenitors. In vivo cell tracking demonstrates that KA″ cells are generated from LFP progenitors by both symmetric and asymmetric cell divisions. A photoconvertible reporter of signaling history (PHRESH) reveals distinct temporal profiles of Hh response: LFP progenitors continuously respond to Hh, while KA″ cells lose Hh response upon differentiation. Hh signaling is required in LFP progenitors for KA″ fate specification, but prolonged Hh signaling interferes with KA″ differentiation. Notch signaling acts permissively to maintain LFP progenitor cells: activation of Notch signaling prevents differentiation, whereas inhibition of Notch signaling results in differentiation of ectopic KA″ cells. These results indicate that neural progenitors depend on Notch signaling to maintain Hh responsiveness and rely on Hh signaling to induce fate identity, whereas proper differentiation depends on the attenuation of both Notch and Hh signaling. During tissue formation, progenitor cells generate both differentiated cells and progenitor cells. It is poorly understood how this balance between self-renewal and differentiation generates the correct number of different cell types. Here, we use zebrafish spinal cord development as a model system to investigate how neural progenitor cells switch from progenitor states to differentiated fates. Combining genetic manipulation and a novel method to study cell signaling in live embryos, our data show that this process requires the dynamic regulation of two signaling pathways: the Notch signaling pathway and the Hedgehog (Hh) signaling pathway. In neural progenitors, Notch signaling maintains the competence of neural progenitors to respond to Hh signaling. In parallel, Hedgehog signaling functions to induce cell fate identity. As cells switch from progenitor states to differentiated states, both Notch and Hh signaling become attenuated. Thus, the dynamic deployment of Notch and Hh signaling controls the renewal and differentiation of progenitor cells.
Collapse
|
29
|
Jung SH, Kim HS, Ryu JH, Gwak JW, Bae YK, Kim CH, Yeo SY. Her4-positive population in the tectum opticum is proliferating neural precursors in the adult zebrafish brain. Mol Cells 2012; 33:627-32. [PMID: 22570150 PMCID: PMC3887756 DOI: 10.1007/s10059-012-0091-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 04/04/2012] [Indexed: 11/25/2022] Open
Abstract
Previous studies have shown that Notch signaling not only regulates the number of early differentiating neurons, but also maintains proliferating neural precursors in the neural tube. Although it is well known that Notch signaling is closely related to the differentiation of adult neural stem cells, none of transgenic zebrafish provides a tool to figure out the relationship between Notch signaling and the differentiation of neural precursors. The goal of this study was to characterize Her4-positive cells by comparing the expression of a fluorescent Her4 reporter in Tg[her4-dRFP] animals with a GFAP reporter in Tg[gfap-GFP] adult zebrafish. BrdU incorporation indicated that dRFP-positive cells were proliferating and a double labeling assay revealed that a significant fraction of the Her4-dRFP positive population was also GFAP-GFP positive. Our observations suggest that a reporter line with Notch-dependent gene expression can provide a tool to examine proliferating neural precursors and/or neuronal/glial precursors in the development of the adult nervous system to examine the model in which Notch signaling maintains proliferating neural precursors in the neural tube.
Collapse
Affiliation(s)
- Seung-Hyun Jung
- Department of Biology and GRAST, Chungnam National University, Daejeon 305-764,
Korea
| | - Hyung-Seok Kim
- Department of Genetic Engineering, Kyungpook National University, Daegu 702-701,
Korea
| | - Jae-Ho Ryu
- Department of Biology and GRAST, Chungnam National University, Daejeon 305-764,
Korea
| | - Jung-Woo Gwak
- Department of Biotechnology, Division of Applied Chemistry and Biotechnology, Hanbat National University, Daejeon 305-719,
Korea
| | - Young-Ki Bae
- Research Institute, National Cancer Center, Goyang 410-769,
Korea
| | - Cheol-Hee Kim
- Department of Biology and GRAST, Chungnam National University, Daejeon 305-764,
Korea
| | - Sang-Yeob Yeo
- Department of Biotechnology, Division of Applied Chemistry and Biotechnology, Hanbat National University, Daejeon 305-719,
Korea
| |
Collapse
|
30
|
Notch signaling controls generation of motor neurons in the lesioned spinal cord of adult zebrafish. J Neurosci 2012; 32:3245-52. [PMID: 22378895 DOI: 10.1523/jneurosci.6398-11.2012] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In mammals, increased Notch signaling is held partly responsible for a lack of neurogenesis after a spinal injury. However, this is difficult to test in an essentially nonregenerating system. We show that in adult zebrafish, which exhibit lesion-induced neurogenesis, e.g., of motor neurons, the Notch pathway is also reactivated. Although apparently compatible with neuronal regeneration in zebrafish, forced activity of the pathway significantly decreased progenitor proliferation and motor neuron generation. Conversely, pharmacological inhibition of the pathway increased proliferation and motor neuron numbers. This demonstrates that Notch is a negative signal for regenerative neurogenesis, and, importantly, that spinal motor neuron regeneration can be augmented in an adult vertebrate by inhibiting Notch signaling.
Collapse
|
31
|
Rabadán MA, Cayuso J, Le Dréau G, Cruz C, Barzi M, Pons S, Briscoe J, Martí E. Jagged2 controls the generation of motor neuron and oligodendrocyte progenitors in the ventral spinal cord. Cell Death Differ 2012; 19:209-19. [PMID: 21720386 PMCID: PMC3263496 DOI: 10.1038/cdd.2011.84] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 05/12/2011] [Accepted: 05/13/2011] [Indexed: 12/20/2022] Open
Abstract
In the developing spinal cord, motor neurons (MNs) and oligodendrocytes arise sequentially from a common pool of progenitors. However, the genetic network responsible for this neurogenesis to gliogenesis switch is largely unknown. A transcriptome analysis identified the Notch ligand Jagged2 (JAG2) as a Sonic hedgehog-regulated factor transiently expressed in MN progenitors (pMNs). In vivo loss- and gain-of-function experiments show that JAG2 schedules the differentiation of the pMN progenitors. At early developmental stages, Olig2 expressing pMN progenitors that enter the differentiation pathway exclusively generate MNs. At these times, the activation of the Notch pathway by JAG2 maintains selected pMN progenitors in an undifferentiated state by two mechanisms; first it inhibits MN generation by reducing Olig2 proteins levels, and second it directly inhibits the premature generation of oligodendrocyte progenitors (OLPs) by maintaining high levels of Hes5. Later, extinction of JAG2 from the pMN results in the loss of Hes5 expression, heralding the gliogenic phase of pMN progenitors. Strikingly, downregulation of JAG2 in pMN progenitors is sufficient to promote the precocious generation of OLPs. Together these data provide evidence that JAG2 is a key regulator of the timely and ordered generation of two of the defining cell types in the spinal cord, MNs and OLPs.
Collapse
Affiliation(s)
- M A Rabadán
- Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, Barcelona, Spain
| | - J Cayuso
- Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, Barcelona, Spain
| | - G Le Dréau
- Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, Barcelona, Spain
| | - C Cruz
- National Institute for Medical Research, The Ridgeway, Mill Hill, London, UK
| | - M Barzi
- Instituto de Investigaciones Biomédicas de Barcelona, CSIC-IDIBAPS, Barcelona, Spain
| | - S Pons
- Instituto de Investigaciones Biomédicas de Barcelona, CSIC-IDIBAPS, Barcelona, Spain
| | - J Briscoe
- National Institute for Medical Research, The Ridgeway, Mill Hill, London, UK
| | - E Martí
- Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, Barcelona, Spain
| |
Collapse
|
32
|
Amlin-Van Schaick JC, Kim S, DiFabio C, Lee MH, Broman KW, Reilly KM. Arlm1 is a male-specific modifier of astrocytoma resistance on mouse Chr 12. Neuro Oncol 2012; 14:160-74. [PMID: 22234937 PMCID: PMC3266387 DOI: 10.1093/neuonc/nor206] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 10/21/2011] [Indexed: 12/18/2022] Open
Abstract
While many cancers show a sex bias, the genetic basis and molecular mechanisms underlying sex bias are not always clear. Astrocytoma and glioblastoma show male predominance in humans. We have shown previously that glial tumors forming in the Nf1-/+; Trp53-/+cis (NPcis) mouse model also show a sex bias in some genetic contexts. Using cross-species comparisons we have identified candidate male-specific modifiers of astrocytoma/glioblastoma. Linkage analysis of B6X(B6X129)-NPcis mice identifies a modifier of astrocytoma resistance specific to males, named Arlm1, on distal mouse Chr 12. Arlm1 is syntenic to human Chr 7p15, 7p21, 7q36, and 14q32 regions that are altered in human glioblastoma. A subset of these genes shows male-specific correlations to glioblastoma patient survival time and represents strong candidates for the Arlm1 modifier gene. Identification of male-specific modifier genes will lead to a better understanding of the molecular basis of male predominance in astrocytoma and glioblastoma.
Collapse
Affiliation(s)
| | | | | | | | | | - Karlyne M. Reilly
- Mouse Cancer Genetics Program, National Cancer Institute, Frederick, Maryland (J.C.A.-V.S., C.D.F., M.-H.L., K.M.R.); Institute for Biomedical Sciences, George Washington University, Washington, DC (J.C.A.-V.S.); Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin (S.K., K.W.B.)
| |
Collapse
|
33
|
Skaggs K, Martin DM, Novitch BG. Regulation of spinal interneuron development by the Olig-related protein Bhlhb5 and Notch signaling. Development 2011; 138:3199-211. [PMID: 21750031 DOI: 10.1242/dev.057281] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The neural circuits that control motor activities depend on the spatially and temporally ordered generation of distinct classes of spinal interneurons. Despite the importance of these interneurons, the mechanisms underlying their genesis are poorly understood. Here, we demonstrate that the Olig-related transcription factor Bhlhb5 (recently renamed Bhlhe22) plays two central roles in this process. Our findings suggest that Bhlhb5 repressor activity acts downstream of retinoid signaling and homeodomain proteins to promote the formation of dI6, V1 and V2 interneuron progenitors and their differentiated progeny. In addition, Bhlhb5 is required to organize the spatially restricted expression of the Notch ligands and Fringe proteins that both elicit the formation of the interneuron populations that arise adjacent to Bhlhb5(+) cells and influence the global pattern of neuronal differentiation. Through these actions, Bhlhb5 helps transform the spatial information established by morphogen signaling into local cell-cell interactions associated with Notch signaling that control the progression of neurogenesis and extend neuronal diversity within the developing spinal cord.
Collapse
Affiliation(s)
- Kaia Skaggs
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | |
Collapse
|
34
|
Cytokines regulate neuronal gene expression: Differential effects of Th1, Th2 and monocyte/macrophage cytokines. J Neuroimmunol 2011; 238:19-33. [DOI: 10.1016/j.jneuroim.2011.06.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 06/16/2011] [Accepted: 06/17/2011] [Indexed: 12/19/2022]
|
35
|
Hortopan GA, Baraban SC. Aberrant expression of genes necessary for neuronal development and Notch signaling in an epileptic mind bomb zebrafish. Dev Dyn 2011; 240:1964-76. [PMID: 21688347 PMCID: PMC3137702 DOI: 10.1002/dvdy.22680] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2011] [Indexed: 11/11/2022] Open
Abstract
Mutation within an ubiquitin E3 ligase gene can lead to a failure in Notch signaling, excessive neurons, and depletion of neural progenitor cells in mind bomb mutants. Using mib(hi904) zebrafish, we reported seizures and a down-regulation of γ-aminobutyric acid (GABA) signaling pathway genes. A transcriptome analysis also identified differential expression pattern of genes related to Notch signaling and neurodevelopment. Here, we selected nine of these genes (her4.2, hes5, bhlhb5, hoxa5a, hoxb5b, dmbx1a, dbx1a, nxph1, and plxnd1) and performed a more thorough analysis of expression using conventional polymerase chain reaction, real-time polymerase chain reaction and in situ hybridization. Transgenic reporter fish (Gfap:GFP and Dlx5a-6a:GFP) were used to assess early brain morphology in vivo. Down-regulation of many of these genes was prominent throughout key structures of the developing mib(hi904) zebrafish brain including, but not limited to, the pallium, ventral thalamus, and optic tectum. Brain expression of Dlx5a-6a and Gfap was also reduced. In conclusion, these expression studies indicate a general down-regulation of Notch signaling genes necessary for proper brain development and suggest that these mutant fish could provide valuable insights into neurological conditions, such as Angelman syndrome, associated with ubiquitin E3 ligase mutation.
Collapse
Affiliation(s)
- Gabriela A. Hortopan
- Epilepsy Research Laboratory, Department of Neurological Surgery, University of California, San Francisco, San Francisco, California 94143
| | - Scott C. Baraban
- Epilepsy Research Laboratory, Department of Neurological Surgery, University of California, San Francisco, San Francisco, California 94143
| |
Collapse
|
36
|
Li H, Paes de Faria J, Andrew P, Nitarska J, Richardson W. Phosphorylation regulates OLIG2 cofactor choice and the motor neuron-oligodendrocyte fate switch. Neuron 2011; 69:918-29. [PMID: 21382552 PMCID: PMC3093612 DOI: 10.1016/j.neuron.2011.01.030] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2011] [Indexed: 01/22/2023]
Abstract
A fundamental feature of central nervous system development is that neurons are generated before glia. In the embryonic spinal cord, for example, a group of neuroepithelial stem cells (NSCs) generates motor neurons (MNs), before switching abruptly to oligodendrocyte precursors (OLPs). We asked how transcription factor OLIG2 participates in this MN-OLP fate switch. We found that Serine 147 in the helix-loop-helix domain of OLIG2 was phosphorylated during MN production and dephosphorylated at the onset of OLP genesis. Mutating Serine 147 to Alanine (S147A) abolished MN production without preventing OLP production in transgenic mice, chicks, or cultured P19 cells. We conclude that S147 phosphorylation, possibly by protein kinase A, is required for MN but not OLP genesis and propose that dephosphorylation triggers the MN-OLP switch. Wild-type OLIG2 forms stable homodimers, whereas mutant (unphosphorylated) OLIG2(S147A) prefers to form heterodimers with Neurogenin 2 or other bHLH partners, suggesting a molecular basis for the switch.
Collapse
Affiliation(s)
- Huiliang Li
- Wolfson Institute for Biomedical Research and Research Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Joana Paes de Faria
- Wolfson Institute for Biomedical Research and Research Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Paul Andrew
- Wolfson Institute for Biomedical Research and Research Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Justyna Nitarska
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - William D. Richardson
- Wolfson Institute for Biomedical Research and Research Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
37
|
Ahn HJ, Park Y, Kim S, Park HC, Seo SK, Yeo SY, Geum D. The expression profile and function of Satb2 in zebrafish embryonic development. Mol Cells 2010; 30:377-82. [PMID: 20814748 DOI: 10.1007/s10059-010-0128-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Revised: 07/17/2010] [Accepted: 07/19/2010] [Indexed: 12/16/2022] Open
Abstract
The present study shows the expression profile and function of the homeobox gene, satb2 during zebrafish embryonic development. Satb2 was ubiquitously expressed from the 1 cell stage to the 10-somite stage in zebrafish embryos. Satb2 showed stage-specific expression profiles such as in the pronephric duct at 24 hpf, the branchial arches at 36 hpf, and the ganglion cell layer of the retina and fins at 48 hpf. Additionally, satb2 knockdown embryos were arrested at 50-60% epiboly, and transplantation experiments with satb2 knockdown cells showed migration defects. Interestingly, satb2 knockdown cells also exhibited down-regulation of dynamin II and VAMP4, which are involved in exocytosis and endocytosis, respectively. Furthermore, satb2 knockdown cells have a disorganized actin distribution and an underdeveloped external yolk syncytial layer, both of which are involved in epiboly. These results suggest that satb2 has a functional role in epiboly. This role may potentially be the regulation of endo-exocytic vesicle transport-dependent cell migration and/or the regulation of the development of the yolk syncytial layer.
Collapse
Affiliation(s)
- Hyun-Jong Ahn
- Department of Microbiology, Kyung Hee University Medical School, Seoul, 130-701, Korea
| | | | | | | | | | | | | |
Collapse
|
38
|
Yang L, Rastegar S, Strähle U. Regulatory interactions specifying Kolmer-Agduhr interneurons. Development 2010; 137:2713-22. [DOI: 10.1242/dev.048470] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In the zebrafish spinal cord, two classes of neurons develop from the lateral floor plate: Kolmer-Agduhr′ (KA′) and V3 interneurons. We show here that the differentiation of the correct number of KA′ cells depends on the activity of the homeobox transcription factor Nkx2.9. This factor acts in concert with Nkx2.2a and Nkx2.2b. These factors are also required for the expression of the zinc-finger transcription factor Gata2 in the lateral floor plate. In turn, Gata2 is necessary for expression of the basic helix-loop-helix transcription factor Tal2 that acts upstream of the GABA-synthesizing enzyme glutamic acid decarboxylase 67 gene (gad67) in KA′ cells. Expression of the transcription factor Sim1, which marks the V3 interneurons in the lateral floor plate, depends also on the three Nkx2 factors. sim1 expression does not require, however, gata2 and tal2. KA′ cells of the lateral floor plate and the KA′ cells located more dorsally in the spinal cord share expression of transcription factors. The functional connections between the different regulatory genes, however, differ in the two GABAergic cell types: although gata2 and tal2 are expressed in KA′ cells, they are dispensable for gad67 expression in these cells. Instead, olig2 and gata3 are required for the differentiation of gad67-expressing KA′ cells. This suggests that the layout of regulatory networks is crucially dependent on the lineage that differs between KA′ and KA′ cells.
Collapse
Affiliation(s)
- Lixin Yang
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Postfach 3640, 76021 Karlsruhe, Germany
| | - Sepand Rastegar
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Postfach 3640, 76021 Karlsruhe, Germany
| | - Uwe Strähle
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Postfach 3640, 76021 Karlsruhe, Germany
| |
Collapse
|
39
|
Gwak JW, Kong HJ, Bae YK, Kim MJ, Lee J, Park JH, Yeo SY. Proliferating neural progenitors in the developing CNS of zebrafish require Jagged2 and Jagged1b. Mol Cells 2010; 30:155-9. [PMID: 20680491 DOI: 10.1007/s10059-010-0101-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Accepted: 04/27/2010] [Indexed: 01/26/2023] Open
Abstract
In the central nervous system (CNS), giving rise to the diversity and the complexity of neurons is the spatial and temporal differentiation of neural stem cells and/or neural precursors. Here, we investigated the role of Jagged-mediated Notch signaling in the maintenance and differentiation of progenitor cells during late neurogenesis by analyzing the expression patterns of zebrafish jagged homologues, and by injecting their morpholinos. Expression of both jagged2 and jagged1b mRNA in the CNS suggested that they might be involved in control of differentiating neural progenitors in which they are involved later in development. In Jagged2 and Jagged1b knock-down embryos, the overall rate of cell division dramatically decreased, and the ectopic VeMe neurons were generated. The results suggest that Jagged-Notch signaling plays a critical role in the maintenance of proliferating neural precursors, and that the generation of late-born neurons, especially VeMe neurons, is regulated by the interplay between Jagged2 and Jagged1b.
Collapse
Affiliation(s)
- Jung-Woo Gwak
- Department of Biotechnology, Division of Applied Chemistry and Biotechnology, Hanbat National University, Daejeon 305-719, Korea
| | | | | | | | | | | | | |
Collapse
|
40
|
Chapouton P, Skupien P, Hesl B, Coolen M, Moore JC, Madelaine R, Kremmer E, Faus-Kessler T, Blader P, Lawson ND, Bally-Cuif L. Notch activity levels control the balance between quiescence and recruitment of adult neural stem cells. J Neurosci 2010; 30:7961-74. [PMID: 20534844 PMCID: PMC6632678 DOI: 10.1523/jneurosci.6170-09.2010] [Citation(s) in RCA: 196] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Accepted: 04/09/2010] [Indexed: 12/19/2022] Open
Abstract
The limited generation of neurons during adulthood is controlled by a balance between quiescence and recruitment of neural stem cells (NSCs). We use here the germinal zone of the zebrafish adult telencephalon to examine how the frequency of NSC divisions is regulated. We show, using several in vivo techniques, that progenitors transit back and forth between the quiescent and dividing state, according to varying levels of Notch activity: Notch induction drives progenitors into quiescence, whereas blocking Notch massively reinitiates NSC division and subsequent commitment toward becoming neurons. Notch activation appears predominantly triggered by newly recruited progenitors onto their neighbors, suggesting an involvement of Notch in a self-limiting mechanism, once neurogenesis is started. These results identify for the first time a lateral inhibition-like mechanism in the context of adult neurogenesis and suggest that the equilibrium between quiescence and neurogenesis in the adult brain is controlled by fluctuations of Notch activity, thereby regulating the amount of adult-born neurons.
Collapse
Affiliation(s)
- Prisca Chapouton
- Zebrafish Neurogenetics Department, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Center for Integrated Protein Science (Munich), Institute of Developmental Genetics, Technical University Munich, D-85764 Neuherberg, Germany
| | - Paulina Skupien
- Zebrafish Neurogenetics Department, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Center for Integrated Protein Science (Munich), Institute of Developmental Genetics, Technical University Munich, D-85764 Neuherberg, Germany
| | - Birgit Hesl
- Zebrafish Neurogenetics Department, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Center for Integrated Protein Science (Munich), Institute of Developmental Genetics, Technical University Munich, D-85764 Neuherberg, Germany
| | - Marion Coolen
- Zebrafish Neurogenetics Department, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Center for Integrated Protein Science (Munich), Institute of Developmental Genetics, Technical University Munich, D-85764 Neuherberg, Germany
- Zebrafish Neurogenetics Group, Laboratory of Neurobiology and Development, CNRS, Unité Propre de Recherche 3294, Institute of Neurobiology Alfred Fessard, F-91198 Gif-sur-Yvette, France
| | - John C. Moore
- Program in Gene Function and Expression, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Romain Madelaine
- Université de Toulouse, Université Paul Sabatier, Centre de Biologie du Développement, F-31062 Toulouse, France
- Centre National de la Recherche Scientifique (CNRS), Centre de Biologie du Développement, Unité Mixte de Recherche 5547, F-31062 Toulouse, France
| | - Elizabeth Kremmer
- Institute of Molecular Immunology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-81377 Munich, Germany
| | - Theresa Faus-Kessler
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany, and
| | - Patrick Blader
- Université de Toulouse, Université Paul Sabatier, Centre de Biologie du Développement, F-31062 Toulouse, France
- Centre National de la Recherche Scientifique (CNRS), Centre de Biologie du Développement, Unité Mixte de Recherche 5547, F-31062 Toulouse, France
| | - Nathan D. Lawson
- Program in Gene Function and Expression, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Laure Bally-Cuif
- Zebrafish Neurogenetics Department, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Center for Integrated Protein Science (Munich), Institute of Developmental Genetics, Technical University Munich, D-85764 Neuherberg, Germany
- Zebrafish Neurogenetics Group, Laboratory of Neurobiology and Development, CNRS, Unité Propre de Recherche 3294, Institute of Neurobiology Alfred Fessard, F-91198 Gif-sur-Yvette, France
| |
Collapse
|
41
|
Gray SD, Dale JK. Notch signalling regulates the contribution of progenitor cells from the chick Hensen's node to the floor plate and notochord. Development 2010; 137:561-8. [PMID: 20110321 DOI: 10.1242/dev.041608] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Hensen's node of the chick embryo contains multipotent self-renewing progenitor cells that can contribute to either the floor plate or the notochord. Floor plate cells are a population of epithelial cells that lie at the ventral midline of the developing neural tube, whereas the notochord is a rod of axial mesoderm that lies directly beneath the floor plate. These two tissues serve as a source of a potent signalling morphogen, sonic hedgehog (Shh), which patterns the dorsoventral axis of the neural tube. We show, through both gain- and loss-of-function approaches, that Notch signalling promotes the contribution of chick axial progenitor cells to the floor plate and inhibits contribution to the notochord. Thus, we propose that Notch regulates the allocation of appropriate numbers of progenitor cells from Hensen's node of the chick embryo to the notochord and the floor plate.
Collapse
Affiliation(s)
- Shona D Gray
- Division of Cell and Developmental Biology, College of Life Sciences, University of Dundee, Dundee, Scotland, UK
| | | |
Collapse
|
42
|
Abstract
For more than a decade, the zebrafish has proven to be an excellent model organism to investigate the mechanisms of neurogenesis during development. The often cited advantages, namely external development, genetic, and optical accessibility, have permitted direct examination and experimental manipulations of neurogenesis during development. Recent studies have begun to investigate adult neurogenesis, taking advantage of its widespread occurrence in the mature zebrafish brain to investigate the mechanisms underlying neural stem cell maintenance and recruitment. Here we provide a comprehensive overview of the tools and techniques available to study neurogenesis in zebrafish both during development and in adulthood. As useful resources, we provide tables of available molecular markers, transgenic, and mutant lines. We further provide optimized protocols for studying neurogenesis in the adult zebrafish brain, including in situ hybridization, immunohistochemistry, in vivo lipofection and electroporation methods to deliver expression constructs, administration of bromodeoxyuridine (BrdU), and finally slice cultures. These currently available tools have put zebrafish on par with other model organisms used to investigate neurogenesis.
Collapse
Affiliation(s)
- Prisca Chapouton
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | | |
Collapse
|
43
|
Notch independent signalling mediates Schwann cell-like differentiation of Adipose Derived Stem Cells. Neurosci Lett 2009; 467:164-8. [DOI: 10.1016/j.neulet.2009.10.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Revised: 10/05/2009] [Accepted: 10/07/2009] [Indexed: 12/19/2022]
|
44
|
Yeo SY. Zebrafish CiA interneurons are late-born primary neurons. Neurosci Lett 2009; 466:131-4. [PMID: 19800937 DOI: 10.1016/j.neulet.2009.09.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Revised: 08/25/2009] [Accepted: 09/23/2009] [Indexed: 11/28/2022]
Abstract
Pax2 is a neural-related transcription factor downstream of Notch signaling and is expressed in the developing spinal cord of zebrafish, including in CiA interneurons. However, the characteristics of pax2-positive neurons are largely unknown. The goal of this study was to characterize Pax2-positive neurons by examining their expression in embryos in which Notch function had been knocked down by mutation or injection of a morpholino or mRNA. I found that Pax2-positive CiA interneurons were late-differentiating primary neurons. pax2.1 was expressed in CoPA commissural neurons and CiA interneurons at 26 hpf. The number of pax2.1-positive cells increased in mind bomb mutant embryos or embryos injected with Su(H)1-MO, but not in cells injected with Xenopus Delta or Delta(stu) mRNA. These observations imply that Notch signaling plays a role in regulating the number of CiA neurons by preventing uncommitted precursors from acquiring a neuronal fate during vertebrate development.
Collapse
Affiliation(s)
- Sang-Yeob Yeo
- Department of Biotechnology, Division of Applied Chemistry & Biotechnology, Hanbat National University, SAN16-1, DuckMyoung-Dong, Yuseong-Gu, Daejeon 305-719, Republic of Korea.
| |
Collapse
|
45
|
Rocha SF, Lopes SS, Gossler A, Henrique D. Dll1 and Dll4 function sequentially in the retina and pV2 domain of the spinal cord to regulate neurogenesis and create cell diversity. Dev Biol 2009; 328:54-65. [PMID: 19389377 DOI: 10.1016/j.ydbio.2009.01.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2008] [Revised: 12/26/2008] [Accepted: 01/05/2009] [Indexed: 12/11/2022]
Abstract
Signalling mediated by Notch receptors is known to have multiple functions during vertebrate neural development, regulating processes like progenitor differentiation and cell type diversification. Various Notch ligands are expressed in the developing nervous system and their activities might contribute to this multiplicity of functions. Here, we show that two Delta-like genes, Dll1 and Dll4, are sequentially expressed in differentiating neurons of the embryonic mouse retina and spinal cord's pV2 domain, with Dll1 starting to be expressed before Dll4. Analysis of Dll1 mutants reveals this gene is necessary and sufficient to maintain a pool of progenitors in the embryonic neuroepithelium. Accordingly, in the spinal cord domains where Dll1 is the only expressed Notch ligand, its inactivation leads to an increased rate of neurogenesis and premature differentiation of neural progenitors. In contrast, in the pV2 domain and retina where Dll1 is co-expressed with Dll4, progenitors are not exhausted and cell diversity is maintained. Together, our results support a model where Dll1 and Dll4 are part of a unique genetic circuitry that regulates subsequent steps of neurogenesis in the retina and pV2 domain: while Dll1 serves to prevent the untimely differentiation of neural progenitors, Dll4 might function to generate diversity within the population of differentiating neurons.
Collapse
Affiliation(s)
- Susana Ferreira Rocha
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | | | | | | |
Collapse
|
46
|
So JH, Chun HS, Bae YK, Kim HS, Park YM, Huh TL, Chitnis AB, Kim CH, Yeo SY. Her4 is necessary for establishing peripheral projections of the trigeminal ganglia in zebrafish. Biochem Biophys Res Commun 2008; 379:22-6. [PMID: 19084503 DOI: 10.1016/j.bbrc.2008.11.149] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Accepted: 11/25/2008] [Indexed: 10/21/2022]
Abstract
Transcripts of notch and its target genes have been detected in some differentiating neurons. However, the role of Notch in neuronal differentiation remains poorly defined. Here, we show that a subset of differentiating sensory neurons in the trigeminal ganglia express her4. Expression of her4 requires Notch signaling during neurogenesis but not during differentiation, when peripheral projections of the trigeminal ganglia are established. These projections develop poorly in her4 morphants. While many components of the canonical Notch signaling pathway are not required for late her4 expression or peripheral axon outgrowth in trigeminal neurons, simultaneous knock-down of Notch receptors prevents establishment of these peripheral projections. These observations suggest that Her4 and Notch play a role in peripheral outgrowth of sensory neurons.
Collapse
Affiliation(s)
- Ju-Hoon So
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Batista MF, Jacobstein J, Lewis KE. Zebrafish V2 cells develop into excitatory CiD and Notch signalling dependent inhibitory VeLD interneurons. Dev Biol 2008; 322:263-75. [PMID: 18680739 DOI: 10.1016/j.ydbio.2008.07.015] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2008] [Revised: 06/25/2008] [Accepted: 07/11/2008] [Indexed: 11/26/2022]
Abstract
The vertebrate spinal cord contains distinct classes of cells that form at precise dorsal-ventral locations and express specific combinations of transcription factors. In amniotes, V2 cells develop in the ventral spinal cord, just dorsal to motoneurons. All V2 cells develop from the same progenitor domain and hence are initially molecularly identical. However, as they start to become post-mitotic and differentiate they subdivide into two intermingled molecularly-distinct subpopulations of cells, V2a and V2b cells. Here we show that the molecular identities of V2a and V2b cells are conserved between zebrafish and amniotes. In zebrafish, these two cell types both develop into interneurons with very similar morphologies, but while V2a cells become excitatory Circumferential Descending (CiD) interneurons, V2b cells become inhibitory Ventral Lateral Descending (VeLD) interneurons. In addition, we demonstrate that Notch signalling is required for V2 cells to develop into V2b cells. In the absence of Notch signalling, all V2b cells develop as V2a cells.
Collapse
Affiliation(s)
- Manuel F Batista
- Department of Physiology, Development and Neuroscience, University of Cambridge, Anatomy Building, Downing Street, Cambridge, UK
| | | | | |
Collapse
|
48
|
Cau E, Quillien A, Blader P. Notch resolves mixed neural identities in the zebrafish epiphysis. Development 2008; 135:2391-401. [PMID: 18550717 DOI: 10.1242/dev.013482] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Manipulation of Notch activity alters neuronal subtype identity in vertebrate neuronal lineages. Nonetheless, it remains controversial whether Notch activity diversifies cell fate by regulating the timing of neurogenesis or acts directly in neuronal subtype specification. Here, we address the role of Notch in the zebrafish epiphysis, a simple structure containing only two neural subtypes: projection neurons and photoreceptors. Reducing the activity of the Notch pathway results in an excess of projection neurons at the expense of photoreceptors, as well as an increase in cells retaining a mixed identity. However, although forced activation of the pathway inhibits the projection neuron fate, it does not promote photoreceptor identity. As birthdating experiments show that projection neurons and photoreceptors are born simultaneously, Notch acts directly during neuronal specification rather than by controlling the timing of neurogenesis. Finally, our data suggest that two distinct signals are required for photoreceptor fate specification: one for the induction of the photoreceptor fate and the other, involving Notch, for the inhibition of projection neuron traits. We propose a novel model in which Notch resolves mixed neural identities by repressing an undesired genetic program.
Collapse
Affiliation(s)
- Elise Cau
- Centre de Biologie du Développement, UMR 5547 CNRS/UPS, Université Paul Sabatier Bât. 4R3, Toulouse Cedex 9, France
| | | | | |
Collapse
|
49
|
Nikopoulos GN, Duarte M, Kubu CJ, Bellum S, Friesel R, Maciag T, Prudovsky I, Verdi JM. Soluble Jagged1 Attenuates Lateral Inhibition, Allowing for the Clonal Expansion of Neural Crest Stem Cells. Stem Cells 2007; 25:3133-42. [PMID: 17761753 DOI: 10.1634/stemcells.2007-0327] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The activation of Notch signaling in neural crest stem cells (NCSCs) results in the rapid loss of neurogenic potential and differentiation into glia. We now show that the attenuation of endogenous Notch signaling within expanding NCSC clones by the Notch ligand soluble Jagged1 (sJ1), maintains NCSCs in a clonal self-renewing state in vitro without affecting their sensitivity to instructive differentiation signals observed previously during NCSC self-renewal. sJ1 functions as a competitive inhibitor of Notch signaling to modulate endogenous cell-cell communication to levels sufficient to inhibit neural differentiation but insufficient to instruct gliogenic differentiation. Attenuated Notch signaling promotes the induction and nonclassic release of fibroblast growth factor 1 (FGF1). The functions of sJ1 and FGF1 signaling are complementary, as abrogation of FGF signaling diminishes the ability of sJ1 to promote NCSC expansion, yet the secondary NCSCs maintain the dosage sensitivity of the founder. These results validate and build upon previous studies on the role of Notch signaling in stem cell self-renewal and suggest that the differentiation bias or self-renewal potential of NCSCs is intrinsically linked to the level of endogenous Notch signaling. This should provide a unique opportunity for the expansion of NCSCs ex vivo without altering their differentiation bias for clinical cell replacement or transplant strategies in tissue repair. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- George N Nikopoulos
- Interdisciplinary Program in Molecular Genetics and Cell Biology, University of Maine, Orono, Maine, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Recent Papers on Zebrafish and Other Aquarium Fish Models. Zebrafish 2007. [DOI: 10.1089/zeb.2007.9987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|