1
|
Zhao R, Moore EL, Gogol MM, Unruh JR, Yu Z, Scott AR, Wang Y, Rajendran NK, Trainor PA. Identification and characterization of intermediate states in mammalian neural crest cell epithelial to mesenchymal transition and delamination. eLife 2024; 13:RP92844. [PMID: 38873887 PMCID: PMC11178358 DOI: 10.7554/elife.92844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a cellular process that converts epithelial cells to mesenchymal cells with migratory potential in developmental and pathological processes. Although originally considered a binary event, EMT in cancer progression involves intermediate states between a fully epithelial and a fully mesenchymal phenotype, which are characterized by distinct combinations of epithelial and mesenchymal markers. This phenomenon has been termed epithelial to mesenchymal plasticity (EMP), however, the intermediate states remain poorly described and it's unclear whether they exist during developmental EMT. Neural crest cells (NCC) are an embryonic progenitor cell population that gives rise to numerous cell types and tissues in vertebrates, and their formation and delamination is a classic example of developmental EMT. However, whether intermediate states also exist during NCC EMT and delamination remains unknown. Through single-cell RNA sequencing of mouse embryos, we identified intermediate NCC states based on their transcriptional signature and then spatially defined their locations in situ in the dorsolateral neuroepithelium. Our results illustrate the importance of cell cycle regulation and functional role for the intermediate stage marker Dlc1 in facilitating mammalian cranial NCC delamination and may provide new insights into mechanisms regulating pathological EMP.
Collapse
Affiliation(s)
- Ruonan Zhao
- Stowers Institute for Medical ResearchKansas CityUnited States
- Department of Anatomy and Cell Biology, University of Kansas Medical CenterKansas CityUnited States
| | - Emma L Moore
- Stowers Institute for Medical ResearchKansas CityUnited States
| | | | - Jay R Unruh
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Zulin Yu
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Allison R Scott
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Yan Wang
- Stowers Institute for Medical ResearchKansas CityUnited States
| | | | - Paul A Trainor
- Stowers Institute for Medical ResearchKansas CityUnited States
- Department of Anatomy and Cell Biology, University of Kansas Medical CenterKansas CityUnited States
| |
Collapse
|
2
|
Zhao R, Moore EL, Gogol MM, Unruh JR, Yu Z, Scott A, Wang Y, Rajendran NK, Trainor PA. Identification and characterization of intermediate states in mammalian neural crest cell epithelial to mesenchymal transition and delamination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.26.564204. [PMID: 37961316 PMCID: PMC10634855 DOI: 10.1101/2023.10.26.564204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Epithelial to mesenchymal transition (EMT) is a cellular process that converts epithelial cells to mesenchymal cells with migratory potential in both developmental and pathological processes. Although originally considered a binary event, EMT in cancer progression involves intermediate states between a fully epithelial and a fully mesenchymal phenotype, which are characterized by distinct combinations of epithelial and mesenchymal markers. This phenomenon has been termed epithelial to mesenchymal plasticity (EMP), however, the intermediate states remain poorly described and it's unclear whether they exist during developmental EMT. Neural crest cells (NCC) are an embryonic progenitor cell population that gives rise to numerous cell types and tissues in vertebrates, and their formation is a classic example of developmental EMT. An important feature of NCC development is their delamination from the neuroepithelium via EMT, following which NCC migrate throughout the embryo and undergo differentiation. NCC delamination shares similar changes in cellular state and structure with cancer cell invasion. However, whether intermediate states also exist during NCC EMT and delamination remains unknown. Through single cell RNA sequencing, we identified intermediate NCC states based on their transcriptional signature and then spatially defined their locations in situ in the dorsolateral neuroepithelium. Our results illustrate the progressive transcriptional and spatial transitions from premigratory to migratory cranial NCC during EMT and delamination. Of note gene expression and trajectory analysis indicate that distinct intermediate populations of NCC delaminate in either S phase or G2/M phase of the cell cycle, and the importance of cell cycle regulation in facilitating mammalian cranial NCC delamination was confirmed through cell cycle inhibition studies. Additionally, transcriptional knockdown revealed a functional role for the intermediate stage marker Dlc1 in regulating NCC delamination and migration. Overall, our work identifying and characterizing the intermediate cellular states, processes, and molecular signals that regulate mammalian NCC EMT and delamination furthers our understanding of developmental EMP and may provide new insights into mechanisms regulating pathological EMP.
Collapse
Affiliation(s)
- Ruonan Zhao
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Emma L. Moore
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | - Jay R. Unruh
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Zulin Yu
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Allison Scott
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Yan Wang
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | - Paul A. Trainor
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
3
|
Shirley CA, Chhabra G, Amiri D, Chang H, Ahmad N. Immune escape and metastasis mechanisms in melanoma: breaking down the dichotomy. Front Immunol 2024; 15:1336023. [PMID: 38426087 PMCID: PMC10902921 DOI: 10.3389/fimmu.2024.1336023] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/24/2024] [Indexed: 03/02/2024] Open
Abstract
Melanoma is one of the most lethal neoplasms of the skin. Despite the revolutionary introduction of immune checkpoint inhibitors, metastatic spread, and recurrence remain critical problems in resistant cases. Melanoma employs a multitude of mechanisms to subvert the immune system and successfully metastasize to distant organs. Concerningly, recent research also shows that tumor cells can disseminate early during melanoma progression and enter dormant states, eventually leading to metastases at a future time. Immune escape and metastasis have previously been viewed as separate phenomena; however, accumulating evidence is breaking down this dichotomy. Recent research into the progressive mechanisms of melanoma provides evidence that dedifferentiation similar to classical epithelial to mesenchymal transition (EMT), genes involved in neural crest stem cell maintenance, and hypoxia/acidosis, are important factors simultaneously involved in immune escape and metastasis. The likeness between EMT and early dissemination, and differences, also become apparent in these contexts. Detailed knowledge of the mechanisms behind "dual drivers" simultaneously promoting metastatically inclined and immunosuppressive environments can yield novel strategies effective in disabling multiple facets of melanoma progression. Furthermore, understanding progression through these drivers may provide insight towards novel treatments capable of preventing recurrence arising from dormant dissemination or improving immunotherapy outcomes.
Collapse
Affiliation(s)
- Carl A. Shirley
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
| | - Gagan Chhabra
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
| | - Deeba Amiri
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
| | - Hao Chang
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
- William S. Middleton Memorial Veterans Hospital, Madison, WI, United States
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
- William S. Middleton Memorial Veterans Hospital, Madison, WI, United States
| |
Collapse
|
4
|
Benítez-Burraco A, Uriagereka J, Nataf S. The genomic landscape of mammal domestication might be orchestrated by selected transcription factors regulating brain and craniofacial development. Dev Genes Evol 2023; 233:123-135. [PMID: 37552321 PMCID: PMC10746608 DOI: 10.1007/s00427-023-00709-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 07/27/2023] [Indexed: 08/09/2023]
Abstract
Domestication transforms once wild animals into tamed animals that can be then exploited by humans. The process entails modifications in the body, cognition, and behavior that are essentially driven by differences in gene expression patterns. Although genetic and epigenetic mechanisms were shown to underlie such differences, less is known about the role exerted by trans-regulatory molecules, notably transcription factors (TFs) in domestication. In this paper, we conducted extensive in silico analyses aimed to clarify the TF landscape of mammal domestication. We first searched the literature, so as to establish a large list of genes selected with domestication in mammals. From this list, we selected genes experimentally demonstrated to exhibit TF functions. We also considered TFs displaying a statistically significant number of targets among the entire list of (domestication) selected genes. This workflow allowed us to identify 5 candidate TFs (SOX2, KLF4, MITF, NR3C1, NR3C2) that were further assessed in terms of biochemical and functional properties. We found that such TFs-of-interest related to mammal domestication are all significantly involved in the development of the brain and the craniofacial region, as well as the immune response and lipid metabolism. A ranking strategy, essentially based on a survey of protein-protein interactions datasets, allowed us to identify SOX2 as the main candidate TF involved in domestication-associated evolutionary changes. These findings should help to clarify the molecular mechanics of domestication and are of interest for future studies aimed to understand the behavioral and cognitive changes associated to domestication.
Collapse
Affiliation(s)
- Antonio Benítez-Burraco
- Department of Spanish, Linguistics, and Theory of Literature (Linguistics), Faculty of Philology, University of Seville, Seville, Spain.
- Área de Lingüística General, Departamento de Lengua Española, Lingüística y Teoría de la Literatura, Facultad de Filología, Universidad de Sevilla, C/ Palos de la Frontera s/n., 41007-, Sevilla, España.
| | - Juan Uriagereka
- Department of Linguistics and School of Languages, Literatures & Cultures, University of Maryland, College Park, MD, USA
| | - Serge Nataf
- Stem-cell and Brain Research Institute, 18 avenue de Doyen Lépine, F-69500, Bron, France
- University of Lyon 1, 43 Bd du 11 Novembre 1918, F-69100, Villeurbanne, France
- Bank of Tissues and Cells, Hospices Civils de Lyon, Hôpital Edouard Herriot, Place d'Arsonval, F-69003, Lyon, France
| |
Collapse
|
5
|
Chuang KT, Chiou SS, Hsu SH. Recent Advances in Transcription Factors Biomarkers and Targeted Therapies Focusing on Epithelial-Mesenchymal Transition. Cancers (Basel) 2023; 15:3338. [PMID: 37444447 DOI: 10.3390/cancers15133338] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/07/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Transcription factors involve many proteins in the process of transactivating or transcribing (none-) encoded DNA to initiate and regulate downstream signals, such as RNA polymerase. Their unique characteristic is that they possess specific domains that bind to specific DNA element sequences called enhancer or promoter sequences. Epithelial-mesenchymal transition (EMT) is involved in cancer progression. Many dysregulated transcription factors-such as Myc, SNAIs, Twists, and ZEBs-are key drivers of tumor metastasis through EMT regulation. This review summarizes currently available evidence related to the oncogenic role of classified transcription factors in EMT editing and epigenetic regulation, clarifying the roles of the classified conserved transcription factor family involved in the EMT and how these factors could be used as therapeutic targets in future investigations.
Collapse
Affiliation(s)
- Kai-Ting Chuang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Shyh-Shin Chiou
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center of Applied Genomics, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Shih-Hsien Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Center of Applied Genomics, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
6
|
Chakraborty S, Kopitchinski N, Zuo Z, Eraso A, Awasthi P, Chari R, Mitra A, Tobias IC, Moorthy SD, Dale RK, Mitchell JA, Petros TJ, Rocha PP. Enhancer-promoter interactions can bypass CTCF-mediated boundaries and contribute to phenotypic robustness. Nat Genet 2023; 55:280-290. [PMID: 36717694 PMCID: PMC10758292 DOI: 10.1038/s41588-022-01295-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 12/20/2022] [Indexed: 01/31/2023]
Abstract
How enhancers activate their distal target promoters remains incompletely understood. Here we dissect how CTCF-mediated loops facilitate and restrict such regulatory interactions. Using an allelic series of mouse mutants, we show that CTCF is neither required for the interaction of the Sox2 gene with distal enhancers, nor for its expression. Insertion of various combinations of CTCF motifs, between Sox2 and its distal enhancers, generated boundaries with varying degrees of insulation that directly correlated with reduced transcriptional output. However, in both epiblast and neural tissues, enhancer contacts and transcriptional induction could not be fully abolished, and insertions failed to disrupt implantation and neurogenesis. In contrast, Sox2 expression was undetectable in the anterior foregut of mutants carrying the strongest boundaries, and these animals fully phenocopied loss of SOX2 in this tissue. We propose that enhancer clusters with a high density of regulatory activity can better overcome physical barriers to maintain faithful gene expression and phenotypic robustness.
Collapse
Affiliation(s)
- Shreeta Chakraborty
- Unit on Genome Structure and Regulation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Nina Kopitchinski
- Unit on Genome Structure and Regulation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Zhenyu Zuo
- Unit on Genome Structure and Regulation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Ariel Eraso
- Unit on Genome Structure and Regulation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Parirokh Awasthi
- Laboratory Animal Sciences Program, Frederick National Lab for Cancer Research, Frederick, MD, USA
| | - Raj Chari
- Laboratory Animal Sciences Program, Frederick National Lab for Cancer Research, Frederick, MD, USA
| | - Apratim Mitra
- Bioinformatics and Scientific Programming Core, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Ian C Tobias
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Sakthi D Moorthy
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Ryan K Dale
- Bioinformatics and Scientific Programming Core, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer A Mitchell
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Timothy J Petros
- Unit on Cellular and Molecular Neurodevelopment, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Pedro P Rocha
- Unit on Genome Structure and Regulation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
7
|
Mandalos NP, Dimou A, Gavala MA, Lambraki E, Remboutsika E. Craniofacial Development Is Fine-Tuned by Sox2. Genes (Basel) 2023; 14:genes14020380. [PMID: 36833308 PMCID: PMC9956624 DOI: 10.3390/genes14020380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/06/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
The precise control of neural crest stem cell delamination, migration and differentiation ensures proper craniofacial and head development. Sox2 shapes the ontogeny of the cranial neural crest to ensure precision of the cell flow in the developing head. Here, we review how Sox2 orchestrates signals that control these complex developmental processes.
Collapse
Affiliation(s)
- Nikolaos Panagiotis Mandalos
- University Research Institute of Maternal and Child Health & Precision Medicine, School of Medicine, National and Kapoditrian University of Athens, 115 27 Athens, Greece
- National Cancer Institute, Frederick, MD 21702, USA
| | - Aikaterini Dimou
- University Research Institute of Maternal and Child Health & Precision Medicine, School of Medicine, National and Kapoditrian University of Athens, 115 27 Athens, Greece
- Center for Translational Medicine and the Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Maria Angeliki Gavala
- University Research Institute of Maternal and Child Health & Precision Medicine, School of Medicine, National and Kapoditrian University of Athens, 115 27 Athens, Greece
- National Technical University of Athens, 157 80 Athens, Greece
| | - Efstathia Lambraki
- University Research Institute of Maternal and Child Health & Precision Medicine, School of Medicine, National and Kapoditrian University of Athens, 115 27 Athens, Greece
- Polytechnic School, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Eumorphia Remboutsika
- University Research Institute of Maternal and Child Health & Precision Medicine, School of Medicine, National and Kapoditrian University of Athens, 115 27 Athens, Greece
- Thrivus Institute for Biomedical Science and Technology, Constellations Ave, Accra GT-336-4330, Ghana
- Correspondence:
| |
Collapse
|
8
|
Place E, Manning E, Kim DW, Kinjo A, Nakamura G, Ohyama K. SHH and Notch regulate SOX9+ progenitors to govern arcuate POMC neurogenesis. Front Neurosci 2022; 16:855288. [PMID: 36033614 PMCID: PMC9404380 DOI: 10.3389/fnins.2022.855288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 07/20/2022] [Indexed: 12/05/2022] Open
Abstract
Pro-opiomelanocortin (POMC)-expressing neurons in the hypothalamic arcuate nucleus (ARC) play key roles in feeding and energy homoeostasis, hence their development is of great research interest. As the process of neurogenesis is accompanied by changes in adhesion, polarity, and migration that resemble aspects of epithelial-to-mesenchymal transitions (EMTs), we have characterised the expression and regulation within the prospective ARC of transcription factors with context-dependent abilities to regulate aspects of EMT. Informed by pseudotime meta-analysis of recent scRNA-seq data, we use immunohistochemistry and multiplex in situ hybridisation to show that SOX2, SRY-Box transcription factor 9 (SOX9), PROX1, Islet1 (ISL1), and SOX11 are sequentially expressed over the course of POMC neurogenesis in the embryonic chick. Through pharmacological studies ex vivo, we demonstrate that while inhibiting either sonic hedgehog (SHH) or Notch signalling reduces the number of SOX9+ neural progenitor cells, these treatments lead, respectively, to lesser and greater numbers of differentiating ISL1+/POMC+ neurons. These results are consistent with a model in which SHH promotes the formation of SOX9+ progenitors, and Notch acts to limit their differentiation. Both pathways are also required to maintain normal levels of proliferation and to suppress apoptosis. Together our findings demonstrate that hypothalamic neurogenesis is accompanied by dynamic expression of transcription factors (TFs) that mediate EMTs, and that SHH and Notch signalling converge to regulate hypothalamic cellular homoeostasis.
Collapse
Affiliation(s)
- Elsie Place
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
| | - Elizabeth Manning
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
| | - Dong Won Kim
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Arisa Kinjo
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Go Nakamura
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Kyoji Ohyama
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
9
|
Thomas R, Menon V, Mani R, Pruszak J. Glycan Epitope and Integrin Expression Dynamics Characterize Neural Crest Epithelial-to-Mesenchymal Transition (EMT) in Human Pluripotent Stem Cell Differentiation. Stem Cell Rev Rep 2022; 18:2952-2965. [PMID: 35727432 DOI: 10.1007/s12015-022-10393-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2022] [Indexed: 10/18/2022]
Abstract
The neural crest gives rise to progeny as diverse as peripheral neurons, myelinating cells, cranial muscle, bone and cartilage tissues, and melanocytes. Neural crest derivation encompasses complex morphological change, including epithelial-to-mesenchymal transition (EMT) and migration to the eventual target locations throughout the body. Neural crest cultures derived from stem cells provide an attractive source for developmental studies in human model systems, of immediate biomedical relevance for neurocristopathies, neural cancer biology and regenerative medicine, if only appropriate markers for lineage and cell type definition and quality control criteria were available. Implementing a defined, scalable protocol to generate neural crest cells from embryonic stem cells, we identify stage-defining cluster-of-differentiation (CD) surface markers during human neural crest development in vitro. Acquisition of increasingly mesenchymal phenotype was characterized by absence of neuroepithelial stemness markers (CD15, CD133, CD49f) and by decrease of CD57 and CD24. Increased per-cell-expression of CD29, CD44 and CD73 correlated with established EMT markers as determined by immunofluorescence and immunoblot analysis. The further development towards migratory neural crest was associated with decreased CD24, CD49f (ITGA6) and CD57 (HNK1) versus an enhanced CD49d (ITGA4), CD49e (ITGA5) and CD51/CD61 (ITGAV/ITGB3) expression. Notably, a shift from CD57 to CD51/CD61 was identified as a sensitive surrogate surface indicator of EMT in neural crest in vitro development. The reported changes in glycan epitope and integrin surface expression may prove useful for elucidating neural crest stemness, EMT progression and malignancies.
Collapse
Affiliation(s)
- Ria Thomas
- Emmy Noether-Group for Stem Cell Biology, Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine and Faculty of Biology, University of Freiburg, Freiburg, Germany.,Neuroregeneration Research Institute, McLean Hospital/ Harvard Medical School, Belmont, MB, USA
| | - Vishal Menon
- Emmy Noether-Group for Stem Cell Biology, Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine and Faculty of Biology, University of Freiburg, Freiburg, Germany.,Wellcome Trust/ Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
| | - Rakesh Mani
- Institute of Anatomy and Cell Biology, Salzburg, Paracelsus Medical University (PMU), Salzburg, Austria.,Center of Anatomy and Cell Biology, Salzburg and Nuremberg, Paracelsus Medical University (PMU), Salzburg, Austria
| | - Jan Pruszak
- Neuroregeneration Research Institute, McLean Hospital/ Harvard Medical School, Belmont, MB, USA. .,Institute of Anatomy and Cell Biology, Salzburg, Paracelsus Medical University (PMU), Salzburg, Austria. .,Center of Anatomy and Cell Biology, Salzburg and Nuremberg, Paracelsus Medical University (PMU), Salzburg, Austria.
| |
Collapse
|
10
|
Xuan L, Fu D, Zhen D, Bai D, Yu L, Gong G. Long non-coding RNA Sox2OT promotes coronary microembolization-induced myocardial injury by mediating pyroptosis. ESC Heart Fail 2022; 9:1689-1702. [PMID: 35304834 PMCID: PMC9065873 DOI: 10.1002/ehf2.13814] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 12/06/2021] [Accepted: 01/12/2022] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE As a common complication of coronary microembolization (CME), myocardial injury (MI) implies high mortality. Long non-coding RNAs (lncRNAs) are rarely studied in CME-induced MI. Herein, this study intended to evaluate the role of lncRNA Sox2 overlapping transcript (Sox2OT) in CME-induced MI. METHODS The CME rat models were successfully established by injection of microemboli. Rat cardiac functions and MI were observed by ultrasonic electrocardiogram, HE staining, and HBFP staining. Functional assays were utilized to test the inflammatory responses, oxidative stress, and pyroptosis using reverse transcription quantitative polymerase chain reaction, Western blotting, immunohistochemistry, immunofluorescence, and ELISA. Dual-luciferase reporter gene assay and RNA immunoprecipitation were conducted to clarify the targeting relations between Sox2OT and microRNA (miRNA)-23b and between miR-23b and toll-like receptor 4 (TLR4). RESULTS Rat CME disrupted the cardiac functions and induced inflammatory responses and oxidative stress, and activated the nuclear factor-kappa B (NF-κB) pathway and pyroptosis (all P < 0.05). An NF-κB inhibitor downregulated the NF-κB pathway, reduced pyroptosis, and relieved cardiomyocyte injury and pyroptosis. Compared with the sham group (1.05 ± 0.32), lncRNA Sox2OT level (4.41 ± 0.67) in the CME group was elevated (P < 0.05). Sox2OT acted as a competitive endogenous RNA (ceRNA) of miR-23b to regulate TLR4. Silencing of Sox2OT favoured miR-23b binding to 3'UTR of TLR4 mRNA leading to suppressed TLR4-mediated NFKB signalling and pyroptosis in myocardial tissues harvested from CME rat models. In addition, miR-23b overexpression could supplement the cytosolic miR-23b reserves to target TLR-4 and partially reverse Sox2OT-mediated pyroptosis in LPS-treated H9C2 cells. CONCLUSIONS This study supported that silencing Sox2OT inhibited CME-induced MI by eliminating Sox2OT/miR-23b binding and down-regulating the TLR4/NF-κB pathway. This investigation may provide novel insights for the treatment of CME-induced MI.
Collapse
Affiliation(s)
- Liying Xuan
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, No. 1742 Holin River Street, Tongliao, Inner Mongolia, 028002, China
- Inner Mongolia Key Laboratory, Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, China
| | - Danni Fu
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, No. 1742 Holin River Street, Tongliao, Inner Mongolia, 028002, China
- Inner Mongolia Key Laboratory, Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, China
| | - Dong Zhen
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, No. 1742 Holin River Street, Tongliao, Inner Mongolia, 028002, China
- Inner Mongolia Key Laboratory, Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, China
| | - Dongsong Bai
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, No. 1742 Holin River Street, Tongliao, Inner Mongolia, 028002, China
- Inner Mongolia Key Laboratory, Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, China
| | - Lijun Yu
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, No. 1742 Holin River Street, Tongliao, Inner Mongolia, 028002, China
- Inner Mongolia Key Laboratory, Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, China
| | - Guohua Gong
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, No. 1742 Holin River Street, Tongliao, Inner Mongolia, 028002, China
- Inner Mongolia Key Laboratory, Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, China
- First Medical Clinic, Inner Mongolia University for Nationalities, Tongliao, China
| |
Collapse
|
11
|
Zhao R, Trainor PA. Epithelial to mesenchymal transition during mammalian neural crest cell delamination. Semin Cell Dev Biol 2022; 138:54-67. [PMID: 35277330 DOI: 10.1016/j.semcdb.2022.02.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 02/08/2022] [Accepted: 02/21/2022] [Indexed: 11/18/2022]
Abstract
Epithelial to mesenchymal transition (EMT) is a well-defined cellular process that was discovered in chicken embryos and described as "epithelial to mesenchymal transformation" [1]. During EMT, epithelial cells lose their epithelial features and acquire mesenchymal character with migratory potential. EMT has subsequently been shown to be essential for both developmental and pathological processes including embryo morphogenesis, wound healing, tissue fibrosis and cancer [2]. During the past 5 years, interest and study of EMT especially in cancer biology have increased exponentially due to the implied role of EMT in multiple aspects of malignancy such as cell invasion, survival, stemness, metastasis, therapeutic resistance and tumor heterogeneity [3]. Since the process of EMT in embryogenesis and cancer progression shares similar phenotypic changes, core transcription factors and molecular mechanisms, it has been proposed that the initiation and development of carcinoma could be attributed to abnormal activation of EMT factors usually required for normal embryo development. Therefore, developmental EMT mechanisms, whose timing, location, and tissue origin are strictly regulated, could prove useful for uncovering new insights into the phenotypic changes and corresponding gene regulatory control of EMT under pathological conditions. In this review, we initially provide an overview of the phenotypic and molecular mechanisms involved in EMT and discuss the newly emerging concept of epithelial to mesenchymal plasticity (EMP). Then we focus on our current knowledge of a classic developmental EMT event, neural crest cell (NCC) delamination, highlighting key differences in our understanding of NCC EMT between mammalian and non-mammalian species. Lastly, we highlight available tools and future directions to advance our understanding of mammalian NCC EMT.
Collapse
Affiliation(s)
- Ruonan Zhao
- Stowers Institute for Medical Research, Kansas City, MO, USA; Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, MO, USA; Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
12
|
Liao J, Huang Y, Wang Q, Chen S, Zhang C, Wang D, Lv Z, Zhang X, Wu M, Chen G. Gene regulatory network from cranial neural crest cells to osteoblast differentiation and calvarial bone development. Cell Mol Life Sci 2022; 79:158. [PMID: 35220463 PMCID: PMC11072871 DOI: 10.1007/s00018-022-04208-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/02/2022] [Accepted: 02/14/2022] [Indexed: 11/03/2022]
Abstract
Calvarial bone is one of the most complex sequences of developmental events in embryology, featuring a uniquely transient, pluripotent stem cell-like population known as the cranial neural crest (CNC). The skull is formed through intramembranous ossification with distinct tissue lineages (e.g. neural crest derived frontal bone and mesoderm derived parietal bone). Due to CNC's vast cell fate potential, in response to a series of inductive secreted cues including BMP/TGF-β, Wnt, FGF, Notch, Hedgehog, Hippo and PDGF signaling, CNC enables generations of a diverse spectrum of differentiated cell types in vivo such as osteoblasts and chondrocytes at the craniofacial level. In recent years, since the studies from a genetic mouse model and single-cell sequencing, new discoveries are uncovered upon CNC patterning, differentiation, and the contribution to the development of cranial bones. In this review, we summarized the differences upon the potential gene regulatory network to regulate CNC derived osteogenic potential in mouse and human, and highlighted specific functions of genetic molecules from multiple signaling pathways and the crosstalk, transcription factors and epigenetic factors in orchestrating CNC commitment and differentiation into osteogenic mesenchyme and bone formation. Disorders in gene regulatory network in CNC patterning indicate highly close relevance to clinical birth defects and diseases, providing valuable transgenic mouse models for subsequent discoveries in delineating the underlying molecular mechanisms. We also emphasized the potential regenerative alternative through scientific discoveries from CNC patterning and genetic molecules in interfering with or alleviating clinical disorders or diseases, which will be beneficial for the molecular targets to be integrated for novel therapeutic strategies in the clinic.
Collapse
Affiliation(s)
- Junguang Liao
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Yuping Huang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Qiang Wang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Sisi Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Chenyang Zhang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Dan Wang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Zhengbing Lv
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xingen Zhang
- Department of Orthopedics, Jiaxing Key Laboratory for Minimally Invasive Surgery in Orthopaedics & Skeletal Regenerative Medicine, Zhejiang Rongjun Hospital, Jiaxing, 314001, China
| | - Mengrui Wu
- Institute of Genetics, College of Life Science, Zhejiang University, Hangzhou, 310058, China
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
- Institute of Genetics, College of Life Science, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
13
|
Zhang A, Aslam H, Sharma N, Warmflash A, Fakhouri WD. Conservation of Epithelial-to-Mesenchymal Transition Process in Neural Crest Cells and Metastatic Cancer. Cells Tissues Organs 2021; 210:151-172. [PMID: 34218225 DOI: 10.1159/000516466] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 04/12/2021] [Indexed: 11/19/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a highly conserved cellular process in several species, from worms to humans. EMT plays a fundamental role in early embryogenesis, wound healing, and cancer metastasis. For neural crest cell (NCC) development, EMT typically results in forming a migratory and potent cell population that generates a wide variety of cell and tissue, including cartilage, bone, connective tissue, endocrine cells, neurons, and glia amongst many others. The degree of conservation between the signaling pathways that regulate EMT during development and metastatic cancer (MC) has not been fully established, despite ample studies. This systematic review and meta-analysis dissects the major signaling pathways involved in EMT of NCC development and MC to unravel the similarities and differences. While the FGF, TGFβ/BMP, SHH, and NOTCH pathways have been rigorously investigated in both systems, the EGF, IGF, HIPPO, Factor Receptor Superfamily, and their intracellular signaling cascades need to be the focus of future NCC studies. In general, meta-analyses of the associated signaling pathways show a significant number of overlapping genes (particularly ligands, transcription regulators, and targeted cadherins) involved in each signaling pathway of both systems without stratification by body segments and cancer type. Lack of stratification makes it difficult to meaningfully evaluate the intracellular downstream effectors of each signaling pathway. Finally, pediatric neuroblastoma and melanoma are NCC-derived malignancies, which emphasize the importance of uncovering the EMT events that convert NCC into treatment-resistant malignant cells.
Collapse
Affiliation(s)
- April Zhang
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Hira Aslam
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Neha Sharma
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Aryeh Warmflash
- Department of Biosciences, Rice University, Houston, Texas, USA
| | - Walid D Fakhouri
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, Texas, USA.,Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center, Houston, Texas, USA
| |
Collapse
|
14
|
Fan X, Masamsetti VP, Sun JQ, Engholm-Keller K, Osteil P, Studdert J, Graham ME, Fossat N, Tam PP. TWIST1 and chromatin regulatory proteins interact to guide neural crest cell differentiation. eLife 2021; 10:62873. [PMID: 33554859 PMCID: PMC7968925 DOI: 10.7554/elife.62873] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/05/2021] [Indexed: 12/11/2022] Open
Abstract
Protein interaction is critical molecular regulatory activity underlining cellular functions and precise cell fate choices. Using TWIST1 BioID-proximity-labeling and network propagation analyses, we discovered and characterized a TWIST-chromatin regulatory module (TWIST1-CRM) in the neural crest cells (NCC). Combinatorial perturbation of core members of TWIST1-CRM: TWIST1, CHD7, CHD8, and WHSC1 in cell models and mouse embryos revealed that loss of the function of the regulatory module resulted in abnormal differentiation of NCCs and compromised craniofacial tissue patterning. Following NCC delamination, low level of TWIST1-CRM activity is instrumental to stabilize the early NCC signatures and migratory potential by repressing the neural stem cell programs. High level of TWIST1 module activity at later phases commits the cells to the ectomesenchyme. Our study further revealed the functional interdependency of TWIST1 and potential neurocristopathy factors in NCC development. Shaping the head and face during development relies on a complex ballet of molecular signals that orchestrates the movement and specialization of various groups of cells. In animals with a backbone for example, neural crest cells (NCCs for short) can march long distances from the developing spine to become some of the tissues that form the skull and cartilage but also the pigment cells and nervous system. NCCs mature into specific cell types thanks to a complex array of factors which trigger a precise sequence of binary fate decisions at the right time and place. Amongst these factors, the protein TWIST1 can set up a cascade of genetic events that control how NCCs will ultimately form tissues in the head. To do so, the TWIST1 protein interacts with many other molecular actors, many of which are still unknown. To find some of these partners, Fan et al. studied TWIST1 in the NCCs of mice and cells grown in the lab. The experiments showed that TWIST1 interacted with CHD7, CHD8 and WHSC1, three proteins that help to switch genes on and off, and which contribute to NCCs moving across the head during development. Further work by Fan et al. then revealed that together, these molecular actors are critical for NCCs to form cells that will form facial bones and cartilage, as opposed to becoming neurons. This result helps to show that there is a trade-off between NCCs forming the face or being part of the nervous system. One in three babies born with a birth defect shows anomalies of the head and face: understanding the exact mechanisms by which NCCs contribute to these structures may help to better predict risks for parents, or to develop new approaches for treatment.
Collapse
Affiliation(s)
- Xiaochen Fan
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Sydney, Australia.,The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Sydney, Australia
| | - V Pragathi Masamsetti
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Jane Qj Sun
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Kasper Engholm-Keller
- Synapse Proteomics Group, Children's Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Pierre Osteil
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Joshua Studdert
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Mark E Graham
- Synapse Proteomics Group, Children's Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Nicolas Fossat
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Sydney, Australia.,The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Sydney, Australia
| | - Patrick Pl Tam
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Sydney, Australia.,The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Sydney, Australia
| |
Collapse
|
15
|
Vidal A, Redmer T. Decoding the Role of CD271 in Melanoma. Cancers (Basel) 2020; 12:cancers12092460. [PMID: 32878000 PMCID: PMC7564075 DOI: 10.3390/cancers12092460] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/10/2020] [Accepted: 08/25/2020] [Indexed: 11/26/2022] Open
Abstract
The evolution of melanoma, the most aggressive type of skin cancer, is triggered by driver mutations that are acquired in the coding regions of particularly BRAF (rat fibrosarcoma serine/threonine kinase, isoform B) or NRAS (neuroblastoma-type ras sarcoma virus) in melanocytes. Although driver mutations strongly determine tumor progression, additional factors are likely required and prerequisite for melanoma formation. Melanocytes are formed during vertebrate development in a well-controlled differentiation process of multipotent neural crest stem cells (NCSCs). However, mechanisms determining the properties of melanocytes and melanoma cells are still not well understood. The nerve growth factor receptor CD271 is likewise expressed in melanocytes, melanoma cells and NCSCs and programs the maintenance of a stem-like and migratory phenotype via a comprehensive network of associated genes. Moreover, CD271 regulates phenotype switching, a process that enables the rapid and reversible conversion of proliferative into invasive or non-stem-like states into stem-like states by yet largely unknown mechanisms. Here, we summarize current findings about CD271-associated mechanisms in melanoma cells and illustrate the role of CD271 for melanoma cell migration and metastasis, phenotype-switching, resistance to therapeutic interventions, and the maintenance of an NCSC-like state.
Collapse
|
16
|
Bishop MR, Diaz Perez KK, Sun M, Ho S, Chopra P, Mukhopadhyay N, Hetmanski JB, Taub MA, Moreno-Uribe LM, Valencia-Ramirez LC, Restrepo Muñeton CP, Wehby G, Hecht JT, Deleyiannis F, Weinberg SM, Wu-Chou YH, Chen PK, Brand H, Epstein MP, Ruczinski I, Murray JC, Beaty TH, Feingold E, Lipinski RJ, Cutler DJ, Marazita ML, Leslie EJ. Genome-wide Enrichment of De Novo Coding Mutations in Orofacial Cleft Trios. Am J Hum Genet 2020; 107:124-136. [PMID: 32574564 PMCID: PMC7332647 DOI: 10.1016/j.ajhg.2020.05.018] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/26/2020] [Indexed: 01/05/2023] Open
Abstract
Although de novo mutations (DNMs) are known to increase an individual's risk of congenital defects, DNMs have not been fully explored regarding orofacial clefts (OFCs), one of the most common human birth defects. Therefore, whole-genome sequencing of 756 child-parent trios of European, Colombian, and Taiwanese ancestry was performed to determine the contributions of coding DNMs to an individual's OFC risk. Overall, we identified a significant excess of loss-of-function DNMs in genes highly expressed in craniofacial tissues, as well as genes associated with known autosomal dominant OFC syndromes. This analysis also revealed roles for zinc-finger homeobox domain and SOX2-interacting genes in OFC etiology.
Collapse
Affiliation(s)
- Madison R. Bishop
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kimberly K. Diaz Perez
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Miranda Sun
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA
| | - Samantha Ho
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Pankaj Chopra
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nandita Mukhopadhyay
- Department of Oral Biology, University of Pittsburgh School of Dental Medicine, Pittsburgh, PA 15219, USA
| | - Jacqueline B. Hetmanski
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Margaret A. Taub
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Lina M. Moreno-Uribe
- Department of Orthodontics, College of Dentistry, University of Iowa, Iowa City, IA 52242, USA
| | | | | | - George Wehby
- Department of Health Management and Policy, College of Public Health, University of Iowa, Iowa City, IA 52242, USA
| | - Jacqueline T. Hecht
- Department of Pediatrics, McGovern Medical School and School of Dentistry, UT Health at Houston, Houston, TX 77030, USA
| | | | - Seth M. Weinberg
- Department of Oral Biology, University of Pittsburgh School of Dental Medicine, Pittsburgh, PA 15219, USA,Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15219, USA
| | - Yah Huei Wu-Chou
- Department of Medical Research, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Philip K. Chen
- Craniofacial Centre, Taipei Medical University Hospital and Taipei Medical University, Taipei, Taiwan
| | - Harrison Brand
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Michael P. Epstein
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ingo Ruczinski
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jeffrey C. Murray
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Terri H. Beaty
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Eleanor Feingold
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15219, USA
| | - Robert J. Lipinski
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA
| | - David J. Cutler
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Mary L. Marazita
- Department of Oral Biology, University of Pittsburgh School of Dental Medicine, Pittsburgh, PA 15219, USA,Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15219, USA
| | - Elizabeth J. Leslie
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA,Corresponding author
| |
Collapse
|
17
|
Skip is essential for Notch signaling to induce Sox2 in cerebral arteriovenous malformations. Cell Signal 2020; 68:109537. [PMID: 31927035 DOI: 10.1016/j.cellsig.2020.109537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 11/24/2022]
Abstract
Notch signaling and Sry-box (Sox) family transcriptional factors both play critical roles in endothelial cell (EC) differentiation in vascularization. Recent studies have shown that excessive Notch signaling induces Sox2 to cause cerebral arteriovenous malformations (AVMs). Here, we examine human pulmonary AVMs and find no induction of Sox2. Results of epigenetic studies also show less alteration of Sox2-DNA binding in pulmonary AVMs than in cerebral AVMs. We identify high expression of ski-interacting protein (Skip) in brain ECs, a Notch-associated chromatin-modifying protein that is lacking in lung ECs. Knockdown of Skip abolished Notch-induction of Sox2 in brain ECs, while restoration of Skip in lung ECs enabled Notch-mediated Sox2 induction. The results suggest that Skip is a key factor for induction of Sox2 in cerebral AVMs.
Collapse
|
18
|
Yao J, Wu X, Zhang D, Wang L, Zhang L, Reynolds EX, Hernandez C, Boström KI, Yao Y. Elevated endothelial Sox2 causes lumen disruption and cerebral arteriovenous malformations. J Clin Invest 2019; 129:3121-3133. [PMID: 31232700 DOI: 10.1172/jci125965] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 04/23/2019] [Indexed: 12/14/2022] Open
Abstract
Lumen integrity in vascularization requires fully differentiated endothelial cells (ECs). Here, we report that endothelial-mesenchymal transitions (EndMTs) emerged in ECs of cerebral arteriovenous malformation (AVMs) and caused disruption of the lumen or lumen disorder. We show that excessive Sry-box 2 (Sox2) signaling was responsible for the EndMTs in cerebral AVMs. EC-specific suppression of Sox2 normalized endothelial differentiation and lumen formation and improved the cerebral AVMs. Epigenetic studies showed that induction of Sox2 altered the cerebral-endothelial transcriptional landscape and identified jumonji domain-containing protein 5 (JMJD5) as a direct target of Sox2. Sox2 interacted with JMJD5 to induce EndMTs in cerebral ECs. Furthermore, we utilized a high-throughput system to identify the β-adrenergic antagonist pronethalol as an inhibitor of Sox2 expression. Treatment with pronethalol stabilized endothelial differentiation and lumen formation, which limited the cerebral AVMs.
Collapse
Affiliation(s)
- Jiayi Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Xiuju Wu
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Daoqin Zhang
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Lumin Wang
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Li Zhang
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Eric X Reynolds
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Carlos Hernandez
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Kristina I Boström
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,The Molecular Biology Institute at UCLA, Los Angeles, California, USA
| | - Yucheng Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
19
|
Wu X, Yao J, Wang L, Zhang D, Zhang L, Reynolds EX, Yu T, Boström KI, Yao Y. Crosstalk between BMP and Notch Induces Sox2 in Cerebral Endothelial Cells. Cells 2019; 8:E549. [PMID: 31174355 PMCID: PMC6628192 DOI: 10.3390/cells8060549] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 05/31/2019] [Accepted: 06/05/2019] [Indexed: 12/26/2022] Open
Abstract
Bone morphogenetic protein (BMP) and Notch signaling are critical for endothelial cell (EC) differentiation in vascular development. Recent studies have shown that excess BMP activity induces Notch signaling in cerebral ECs resulting in arteriovenous malformation (AVMs). However, it is unclear how the crosstalk between BMP and Notch signaling affects cerebral EC differentiation at the gene regulatory level. Here, we report that BMP6 activates the activin receptor-like kinase (ALK) 3, a BMP type 1 receptor, to induce Notch1 receptor and Jagged1 and Jagged2 ligands. We show that increased expression of the Notch components alters the transcriptional regulatory complex in the SRY-Box 2 (Sox2) promoter region so as to induce its expression in cerebral ECs. Together, our results identify Sox2 as a direct target of BMP and Notch signaling and provide information on how altered BMP and Notch signaling affects the endothelial transcriptional landscape.
Collapse
Affiliation(s)
- Xiuju Wu
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1679, USA.
| | - Jiayi Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1679, USA.
| | - Lumin Wang
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1679, USA.
- Department of cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.
| | - Daoqin Zhang
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1679, USA.
| | - Li Zhang
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1679, USA.
| | - Eric X Reynolds
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1679, USA.
| | - Tongtong Yu
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1679, USA.
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Kristina I Boström
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1679, USA.
- The Molecular Biology Institute at UCLA, Los Angeles, CA 90095-1570, USA.
| | - Yucheng Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1679, USA.
| |
Collapse
|
20
|
Yao Y, Yao J, Boström KI. SOX Transcription Factors in Endothelial Differentiation and Endothelial-Mesenchymal Transitions. Front Cardiovasc Med 2019; 6:30. [PMID: 30984768 PMCID: PMC6447608 DOI: 10.3389/fcvm.2019.00030] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 03/07/2019] [Indexed: 12/19/2022] Open
Abstract
The SRY (Sex Determining Region Y)-related HMG box of DNA binding proteins, referred to as SOX transcription factors, were first identified as critical regulators of male sex determination but are now known to play an important role in vascular development and disease. SOX7, 17, and 18 are essential in endothelial differentiation and SOX2 has emerged as an essential mediator of endothelial-mesenchymal transitions (EndMTs), a mechanism that enables the endothelium to contribute cells with abnormal cell differentiation to vascular disease such as calcific vasculopathy. In the following paper, we review published information on the SOX transcription factors in endothelial differentiation and hypothesize that SOX2 acts as a mediator of EndMTs that contribute to vascular calcification.
Collapse
Affiliation(s)
- Yucheng Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Jiayi Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Kristina I Boström
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States.,Molecular Biology Institute, UCLA, Los Angeles, CA, United States
| |
Collapse
|
21
|
Mandalos NP, Karampelas I, Saridaki M, McKay RDG, Cohen ML, Remboutsika E. A Role for Sox2 in the Adult Cerebellum. ACTA ACUST UNITED AC 2018; 8. [PMID: 30568848 DOI: 10.4172/2157-7633.1000433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The cerebellum, a derivative of the hindbrain, plays a crucial role in balance and posture as well as in higher cognitive and locomotive processes. Cerebellar development is initiated during the segmental phase of hindbrain formation. Here, we describe the phenotype, of a single surviving adult conditional mouse mutant mouse, in which Sox2 function is ablated in embryonic radial glial cells by means of hGFAP-CRE. The single Sox2RGINV/mosaic adult mutant mouse displays motor disability, microsomia, reduced Central Nervous System (CNS) size and cerebellar defects associated with human genetically related congenital abnormalities.
Collapse
Affiliation(s)
- Nikolaos Panagiotis Mandalos
- National and Kapodistrian University of Athens, School of Medicine, Department of Pediatrics, Athens, Greece.,Stem Cell Biology Laboratory, Biomedical Sciences Research Centre "Alexander Fleming", Vari-Attica, Greece
| | - Ioannis Karampelas
- Department of Neurosurgery, University Hospitals Case Medical Center, Cleveland, OH, USA
| | - Marannia Saridaki
- Stem Cell Biology Laboratory, Biomedical Sciences Research Centre "Alexander Fleming", Vari-Attica, Greece
| | - Ronald D G McKay
- The Lieber Institute for Brain Development, Basic Sciences Division, Johns Hopkins Medical Campus, Baltimore, USA
| | - Mark L Cohen
- Department of Pathology, University Hospitals of Cleveland, Case Western Reserve School of Medicine, Cleveland, OH, USA
| | - Eumorphia Remboutsika
- National and Kapodistrian University of Athens, School of Medicine, Department of Pediatrics, Athens, Greece.,Stem Cell Biology Laboratory, Biomedical Sciences Research Centre "Alexander Fleming", Vari-Attica, Greece.,The Lieber Institute for Brain Development, Basic Sciences Division, Johns Hopkins Medical Campus, Baltimore, USA
| |
Collapse
|
22
|
Komiya Y, Bai Z, Cai N, Lou L, Al-Saadi N, Mezzacappa C, Habas R, Runnels LW. A Nonredundant Role for the TRPM6 Channel in Neural Tube Closure. Sci Rep 2017; 7:15623. [PMID: 29142255 PMCID: PMC5688082 DOI: 10.1038/s41598-017-15855-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 10/19/2017] [Indexed: 01/17/2023] Open
Abstract
In humans, germline mutations in Trpm6 cause autosomal dominant hypomagnesemia with secondary hypocalcemia disorder. Loss of Trpm6 in mice also perturbs cellular magnesium homeostasis but additionally results in early embryonic lethality and neural tube closure defects. To define the mechanisms by which TRPM6 influences neural tube closure, we functionally characterized the role of TRPM6 during early embryogenesis in Xenopus laevis. The expression of Xenopus TRPM6 (XTRPM6) is elevated at the onset of gastrulation and is concentrated in the lateral mesoderm and ectoderm at the neurula stage. Loss of XTRPM6 produced gastrulation and neural tube closure defects. Unlike XTRPM6's close homologue XTRPM7, whose loss interferes with mediolateral intercalation, depletion of XTRPM6 but not XTRPM7 disrupted radial intercalation cell movements. A zinc-influx assay demonstrated that TRPM6 has the potential to constitute functional channels in the absence of TRPM7. The results of our study indicate that XTRPM6 regulates radial intercalation with little or no contribution from XTRPM7 in the region lateral to the neural plate, whereas XTRPM7 is mainly involved in regulating mediolateral intercalation in the medial region of the neural plate. We conclude that both TRPM6 and TRPM7 channels function cooperatively but have distinct and essential roles during neural tube closure.
Collapse
Affiliation(s)
- Yuko Komiya
- Rutgers-Robert Wood Johnson Medical School, Deptartment of Pharmacology, Piscataway, 08854, USA.
| | - Zhiyong Bai
- Rutgers-Robert Wood Johnson Medical School, Deptartment of Pharmacology, Piscataway, 08854, USA
| | - Na Cai
- Rutgers-Robert Wood Johnson Medical School, Deptartment of Pharmacology, Piscataway, 08854, USA
| | - Liping Lou
- Rutgers-Robert Wood Johnson Medical School, Deptartment of Pharmacology, Piscataway, 08854, USA
| | - Namariq Al-Saadi
- Rutgers-Robert Wood Johnson Medical School, Deptartment of Pharmacology, Piscataway, 08854, USA
| | | | - Raymond Habas
- Temple University, Deptartment of Biology, Philadelphia, 19122, USA.
| | - Loren W Runnels
- Rutgers-Robert Wood Johnson Medical School, Deptartment of Pharmacology, Piscataway, 08854, USA.
| |
Collapse
|
23
|
Villalba-Benito L, Torroglosa A, Fernández RM, Ruíz-Ferrer M, Moya-Jiménez MJ, Antiñolo G, Borrego S. Overexpression of DNMT3b target genes during Enteric Nervous System development contribute to the onset of Hirschsprung disease. Sci Rep 2017; 7:6221. [PMID: 28740121 PMCID: PMC5524929 DOI: 10.1038/s41598-017-06539-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 06/09/2017] [Indexed: 12/27/2022] Open
Abstract
Hirschsprung disease (HSCR) is attributed to a failure of neural crest cells (NCCs) to migrate, proliferate, differentiate and/or survive in the bowel wall during embryonic Enteric Nervous System (ENS) development. ENS formation is the result from a specific gene expression pattern regulated by epigenetic events, such DNA methylation by the DNA methyltransferases (DNMTs), among other mechanisms. Specifically, DNMT3b de novo methyltransferase is associated with NCCs development and has been shown to be implicated in ENS formation and in HSCR. Aiming to elucidate the specific mechanism underlying the DNMT3b role in such processes, we have performed a chromatin immunoprecipitation coupled with massively parallel sequencing analysis to identify the DNMT3B target genes in enteric precursor cells (EPCs) from mice. Moreover, the expression patterns of those target genes have been analyzed in human EPCs from HSCR patients in comparison with controls. Additionally, we have carried out a search of rare variants in those genes in a HSCR series. Through this approach we found 9 genes showing a significantly different expression level in both groups. Therefore, those genes may have a role in the proper human ENS formation and a failure in their expression pattern might contribute to this pathology.
Collapse
Affiliation(s)
- Leticia Villalba-Benito
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, 41013, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, 41013, Spain
| | - Ana Torroglosa
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, 41013, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, 41013, Spain
| | - Raquel María Fernández
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, 41013, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, 41013, Spain
| | - Macarena Ruíz-Ferrer
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, 41013, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, 41013, Spain
| | - María José Moya-Jiménez
- Department of Pediatric Surgery, University Hospital Virgen del Rocío, Seville, 41013, Spain
| | - Guillermo Antiñolo
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, 41013, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, 41013, Spain
| | - Salud Borrego
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, 41013, Spain.
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, 41013, Spain.
| |
Collapse
|
24
|
Mandalos NP, Remboutsika E. Sox2: To crest or not to crest? Semin Cell Dev Biol 2016; 63:43-49. [PMID: 27592260 DOI: 10.1016/j.semcdb.2016.08.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 08/29/2016] [Accepted: 08/30/2016] [Indexed: 12/12/2022]
Abstract
Precise control of neural progenitor transformation into neural crest stem cells ensures proper craniofacial and head development. In the neural progenitor pool, SoxB factors play an essential role as cell fate determinants of neural development, whereas during neural crest stem cell formation, Sox2 plays a predominant role as a guardian of the developmental clock that ensures precision of cell flow in the developing head.
Collapse
Affiliation(s)
- Nikolaos Panagiotis Mandalos
- National University of Athens Medical School, Department of Pediatrics, 75 Mikras Asias Str., 115 27, Athens, Greece; Stem Cell Biology Laboratory, Biomedical Sciences Research Centre "Alexander Fleming", 34 Fleming Str., 16672 Vari-Attica, Greece; Adjunct Faculty, The Lieber Institute for Brain Development, Basic Sciences Division, Johns Hopkins Medical Campus, 855 North Wolfe Str., Suite 300, 3rd Floor, Baltimore, MD 21205, USA
| | - Eumorphia Remboutsika
- National University of Athens Medical School, Department of Pediatrics, 75 Mikras Asias Str., 115 27, Athens, Greece; Stem Cell Biology Laboratory, Biomedical Sciences Research Centre "Alexander Fleming", 34 Fleming Str., 16672 Vari-Attica, Greece; Adjunct Faculty, The Lieber Institute for Brain Development, Basic Sciences Division, Johns Hopkins Medical Campus, 855 North Wolfe Str., Suite 300, 3rd Floor, Baltimore, MD 21205, USA.
| |
Collapse
|
25
|
Asad Z, Pandey A, Babu A, Sun Y, Shevade K, Kapoor S, Ullah I, Ranjan S, Scaria V, Bajpai R, Sachidanandan C. Rescue of neural crest-derived phenotypes in a zebrafish CHARGE model by Sox10 downregulation. Hum Mol Genet 2016; 25:3539-3554. [PMID: 27418670 PMCID: PMC5179949 DOI: 10.1093/hmg/ddw198] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 05/27/2016] [Accepted: 06/20/2016] [Indexed: 12/20/2022] Open
Abstract
CHD7 mutations are implicated in a majority of cases of the congenital disorder, CHARGE syndrome. CHARGE, an autosomal dominant syndrome, is known to affect multiple tissues including eye, heart, ear, craniofacial nerves and skeleton and genital organs. Using a morpholino-antisense-oligonucleotide-based zebrafish model for CHARGE syndrome, we uncover a complex spectrum of abnormalities in the neural crest and the crest-derived cell types. We report for the first time, defects in myelinating Schwann cells, enteric neurons and pigment cells in a CHARGE model. We also observe defects in the specification of peripheral neurons and the craniofacial skeleton as previously reported. Chd7 morphants have impaired migration of neural crest cells and deregulation of sox10 expression from the early stages. Knocking down Sox10 in the zebrafish CHARGE model rescued the defects in Schwann cells and craniofacial cartilage. Our zebrafish CHARGE model thus reveals important regulatory roles for Chd7 at multiple points of neural crest development viz., migration, fate choice and differentiation and we suggest that sox10 deregulation is an important driver of the neural crest-derived aspects of Chd7 dependent CHARGE syndrome.
Collapse
Affiliation(s)
- Zainab Asad
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India and
| | - Aditi Pandey
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
| | - Aswini Babu
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India and
| | - Yuhan Sun
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry and Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kaivalya Shevade
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry and Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shruti Kapoor
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India and
| | - Ikram Ullah
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
| | - Shashi Ranjan
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
| | - Vinod Scaria
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India and
| | - Ruchi Bajpai
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry and Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Chetana Sachidanandan
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India and
| |
Collapse
|
26
|
Poulou M, Mandalos NP, Karnavas T, Saridaki M, McKay RDG, Remboutsika E. A "Hit and Run" Approach to Inducible Direct Reprogramming of Astrocytes to Neural Stem Cells. Front Physiol 2016; 7:127. [PMID: 27148066 PMCID: PMC4828628 DOI: 10.3389/fphys.2016.00127] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/22/2016] [Indexed: 01/12/2023] Open
Abstract
Temporal and spatial control of gene expression can be achieved using an inducible system as a fundamental tool for regulated transcription in basic, applied and eventually in clinical research. We describe a novel “hit and run” inducible direct reprogramming approach. In a single step, 2 days post-transfection, transiently transfected Sox2FLAG under the Leu3p-αIPM inducible control (iSox2) triggers the activation of endogenous Sox2, redirecting primary astrocytes into abundant distinct nestin-positive radial glia cells. This technique introduces a unique novel tool for safe, rapid and efficient reprogramming amendable to regenerative medicine.
Collapse
Affiliation(s)
- Maria Poulou
- Stem Cell Biology Laboratory, Biomedical Sciences Research Centre "Alexander Fleming," Vari-Attica, Greece
| | - Nikolaos P Mandalos
- Stem Cell Biology Laboratory, Biomedical Sciences Research Centre "Alexander Fleming,"Vari-Attica, Greece; Choremio Laboratory, Department of Pediatrics, National University of Athens Medical SchoolAthens, Greece
| | - Theodoros Karnavas
- Stem Cell Biology Laboratory, Biomedical Sciences Research Centre "Alexander Fleming,"Vari-Attica, Greece; Choremio Laboratory, Department of Pediatrics, National University of Athens Medical SchoolAthens, Greece
| | - Marannia Saridaki
- Stem Cell Biology Laboratory, Biomedical Sciences Research Centre "Alexander Fleming," Vari-Attica, Greece
| | - Ronald D G McKay
- Basic Sciences Division, The Lieber Institute for Brain Development Baltimore, MD, USA
| | - Eumorphia Remboutsika
- Stem Cell Biology Laboratory, Biomedical Sciences Research Centre "Alexander Fleming,"Vari-Attica, Greece; Choremio Laboratory, Department of Pediatrics, National University of Athens Medical SchoolAthens, Greece; Basic Sciences Division, The Lieber Institute for Brain DevelopmentBaltimore, MD, USA
| |
Collapse
|
27
|
Yao J, Guihard PJ, Blazquez-Medela AM, Guo Y, Moon JH, Jumabay M, Boström KI, Yao Y. Serine Protease Activation Essential for Endothelial-Mesenchymal Transition in Vascular Calcification. Circ Res 2015; 117:758-69. [PMID: 26265629 PMCID: PMC4600461 DOI: 10.1161/circresaha.115.306751] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 08/11/2015] [Indexed: 12/20/2022]
Abstract
RATIONALE Endothelial cells have the ability to undergo endothelial-mesenchymal transitions (EndMTs), by which they acquire a mesenchymal phenotype and stem cell-like characteristics. We previously found that EndMTs occurred in the endothelium deficient in matrix γ-carboxyglutamic acid protein enabling endothelial cells to contribute cells to vascular calcification. However, the mechanism responsible for initiating EndMTs is not fully understood. OBJECTIVE To determine the role of specific serine proteases and sex determining region Y-box 2 (Sox2) in the initiation of EndMTs. METHODS AND RESULTS In this study, we used in vivo and in vitro models of vascular calcification to demonstrate that serine proteases and Sox2 are essential for the initiation of EndMTs in matrix γ-carboxyglutamic acid protein-deficient endothelium. We showed that expression of a group of specific serine proteases was highly induced in endothelial cells at sites of vascular calcification in Mgp null aortas. Treatment with serine protease inhibitors decreased both stem cell marker expression and vascular calcification. In human aortic endothelial cells, this group of serine proteases also induced EndMTs, and the activation of proteases was mediated by Sox2. Knockdown of the serine proteases or Sox2 diminished EndMTs and calcification. Endothelial-specific deletion of Sox2 decreased expression of stem cell markers and aortic calcification in matrix γ-carboxyglutamic acid protein-deficient mice. CONCLUSIONS Our results suggest that Sox2-mediated activation of specific serine proteases is essential for initiating EndMTs, and thus, may provide new therapeutic targets for treating vascular calcification.
Collapse
MESH Headings
- Animals
- Calcinosis
- Calcium-Binding Proteins/genetics
- Calcium-Binding Proteins/metabolism
- Cells, Cultured
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Endothelium, Vascular/ultrastructure
- Enzyme Activation
- Extracellular Matrix Proteins/genetics
- Extracellular Matrix Proteins/metabolism
- Gene Expression Profiling/methods
- Humans
- Immunoblotting
- Kallikreins/genetics
- Kallikreins/metabolism
- Mesoderm/metabolism
- Mesoderm/pathology
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Microscopy, Electron, Scanning
- Microscopy, Electron, Transmission
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Pancreatic Elastase/genetics
- Pancreatic Elastase/metabolism
- RNA Interference
- Reverse Transcriptase Polymerase Chain Reaction
- SOXB1 Transcription Factors/genetics
- SOXB1 Transcription Factors/metabolism
- Serine Endopeptidases/genetics
- Serine Endopeptidases/metabolism
- Twist-Related Protein 1/genetics
- Twist-Related Protein 1/metabolism
- Matrix Gla Protein
Collapse
Affiliation(s)
- Jiayi Yao
- From the Division of Cardiology, David Geffen School of Medicine (J.Y., P.J.G., A.M.B.-M., Y.G., J.H.M., M.J., K.I.B., Y.Y.) and The Molecular Biology Institute (K.I.B.), University of California, Los Angeles; and Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China (J.Y.)
| | - Pierre J Guihard
- From the Division of Cardiology, David Geffen School of Medicine (J.Y., P.J.G., A.M.B.-M., Y.G., J.H.M., M.J., K.I.B., Y.Y.) and The Molecular Biology Institute (K.I.B.), University of California, Los Angeles; and Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China (J.Y.)
| | - Ana M Blazquez-Medela
- From the Division of Cardiology, David Geffen School of Medicine (J.Y., P.J.G., A.M.B.-M., Y.G., J.H.M., M.J., K.I.B., Y.Y.) and The Molecular Biology Institute (K.I.B.), University of California, Los Angeles; and Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China (J.Y.)
| | - Yina Guo
- From the Division of Cardiology, David Geffen School of Medicine (J.Y., P.J.G., A.M.B.-M., Y.G., J.H.M., M.J., K.I.B., Y.Y.) and The Molecular Biology Institute (K.I.B.), University of California, Los Angeles; and Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China (J.Y.)
| | - Jeremiah H Moon
- From the Division of Cardiology, David Geffen School of Medicine (J.Y., P.J.G., A.M.B.-M., Y.G., J.H.M., M.J., K.I.B., Y.Y.) and The Molecular Biology Institute (K.I.B.), University of California, Los Angeles; and Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China (J.Y.)
| | - Medet Jumabay
- From the Division of Cardiology, David Geffen School of Medicine (J.Y., P.J.G., A.M.B.-M., Y.G., J.H.M., M.J., K.I.B., Y.Y.) and The Molecular Biology Institute (K.I.B.), University of California, Los Angeles; and Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China (J.Y.)
| | - Kristina I Boström
- From the Division of Cardiology, David Geffen School of Medicine (J.Y., P.J.G., A.M.B.-M., Y.G., J.H.M., M.J., K.I.B., Y.Y.) and The Molecular Biology Institute (K.I.B.), University of California, Los Angeles; and Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China (J.Y.)
| | - Yucheng Yao
- From the Division of Cardiology, David Geffen School of Medicine (J.Y., P.J.G., A.M.B.-M., Y.G., J.H.M., M.J., K.I.B., Y.Y.) and The Molecular Biology Institute (K.I.B.), University of California, Los Angeles; and Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China (J.Y.).
| |
Collapse
|
28
|
Shahryari A, Jazi MS, Samaei NM, Mowla SJ. Long non-coding RNA SOX2OT: expression signature, splicing patterns, and emerging roles in pluripotency and tumorigenesis. Front Genet 2015; 6:196. [PMID: 26136768 PMCID: PMC4469893 DOI: 10.3389/fgene.2015.00196] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/18/2015] [Indexed: 12/18/2022] Open
Abstract
SOX2 overlapping transcript (SOX2OT) is a long non-coding RNA which harbors one of the major regulators of pluripotency, SOX2 gene, in its intronic region. SOX2OT gene is mapped to human chromosome 3q26.3 (Chr3q26.3) locus and is extended in a high conserved region of over 700 kb. Little is known about the exact role of SOX2OT; however, recent studies have demonstrated a positive role for it in transcription regulation of SOX2 gene. Similar to SOX2, SOX2OT is highly expressed in embryonic stem cells and down-regulated upon the induction of differentiation. SOX2OT is dynamically regulated during the embryogenesis of vertebrates, and delimited to the brain in adult mice and human. Recently, the disregulation of SOX2OT expression and its concomitant expression with SOX2 have become highlighted in some somatic cancers including esophageal squamous cell carcinoma, lung squamous cell carcinoma, and breast cancer. Interestingly, SOX2OT is differentially spliced into multiple mRNA-like transcripts in stem and cancer cells. In this review, we are describing the structural and functional features of SOX2OT, with an emphasis on its expression signature, its splicing patterns and its critical function in the regulation of SOX2 expression during development and tumorigenesis.
Collapse
Affiliation(s)
- Alireza Shahryari
- Stem Cell Research Center, Golestan University of Medical Sciences , Gorgan, Iran
| | - Marie Saghaeian Jazi
- Department of Molecular Medicine, Faculty of Advanced Medical Technologies, Golestan University of Medical Sciences , Gorgan, Iran
| | - Nader M Samaei
- Department of Medical Genetics, Faculty of Advanced Medical Technologies, Golestan University of Medical Sciences , Gorgan, Iran
| | - Seyed J Mowla
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University , Tehran, Iran
| |
Collapse
|
29
|
Tanabe S. Origin of cells and network information. World J Stem Cells 2015; 7:535-540. [PMID: 25914760 PMCID: PMC4404388 DOI: 10.4252/wjsc.v7.i3.535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 01/20/2015] [Accepted: 02/09/2015] [Indexed: 02/06/2023] Open
Abstract
All cells are derived from one cell, and the origin of different cell types is a subject of curiosity. Cells construct life through appropriately timed networks at each stage of development. Communication among cells and intracellular signaling are essential for cell differentiation and for life processes. Cellular molecular networks establish cell diversity and life. The investigation of the regulation of each gene in the genome within the cellular network is therefore of interest. Stem cells produce various cells that are suitable for specific purposes. The dynamics of the information in the cellular network changes as the status of cells is altered. The components of each cell are subject to investigation.
Collapse
Affiliation(s)
- Shihori Tanabe
- Shihori Tanabe, National Institute of Health Sciences, Tokyo 158-8501, Japan
| |
Collapse
|
30
|
Stergiopoulos A, Elkouris M, Politis PK. Prospero-related homeobox 1 (Prox1) at the crossroads of diverse pathways during adult neural fate specification. Front Cell Neurosci 2015; 8:454. [PMID: 25674048 PMCID: PMC4306308 DOI: 10.3389/fncel.2014.00454] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/16/2014] [Indexed: 12/11/2022] Open
Abstract
Over the last decades, adult neurogenesis in the central nervous system (CNS) has emerged as a fundamental process underlying physiology and disease. Recent evidence indicates that the homeobox transcription factor Prox1 is a critical intrinsic regulator of neurogenesis in the embryonic CNS and adult dentate gyrus (DG) of the hippocampus, acting in multiple ways and instructed by extrinsic cues and intrinsic factors. In the embryonic CNS, Prox1 is mechanistically involved in the regulation of proliferation vs. differentiation decisions of neural stem cells (NSCs), promoting cell cycle exit and neuronal differentiation, while inhibiting astrogliogenesis. During the complex differentiation events in adult hippocampal neurogenesis, Prox1 is required for maintenance of intermediate progenitors (IPs), differentiation and maturation of glutamatergic interneurons, as well as specification of DG cell identity over CA3 pyramidal fate. The mechanism by which Prox1 exerts multiple functions involves distinct signaling pathways currently not fully highlighted. In this mini-review, we thoroughly discuss the Prox1-dependent phenotypes and molecular pathways in adult neurogenesis in relation to different upstream signaling cues and cell fate determinants. In addition, we discuss the possibility that Prox1 may act as a cross-talk point between diverse signaling cascades to achieve specific outcomes during adult neurogenesis.
Collapse
Affiliation(s)
- Athanasios Stergiopoulos
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens Athens, Greece
| | - Maximilianos Elkouris
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens Athens, Greece
| | - Panagiotis K Politis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens Athens, Greece
| |
Collapse
|