1
|
Rigney S, York JR, LaBonne C. Krüppel-like factors play essential roles in regulating pluripotency and the formation of neural crest stem cells. Development 2025; 152:dev204634. [PMID: 40292574 PMCID: PMC12070069 DOI: 10.1242/dev.204634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/21/2025] [Indexed: 04/30/2025]
Abstract
The evolution of complex vertebrate body plans was driven by the acquisition of the neural crest, a stem cell population that retains broad, multi-germ layer potential after most embryonic cells have become lineage restricted. We have previously shown that neural crest cells share significant gene regulatory architecture with pluripotent blastula stem cells. Here, we examine the roles that two Krüppel-like Family (Klf) transcription factors, Klf2 and Klf17, play in these cell populations. We found that inhibition of either klf2 or klf17 expanded expression of pluripotency, neural plate border and neural crest factors in neurula stage Xenopus embryos, suggesting that Klf factors regulate the exit from pluripotency and proper establishment of the boundary of the neural crest domain. To gain further insights into the role of Klf factors in the evolution of the neural crest, we examined their expression in sea lamprey, a jawless vertebrate, and show that ectopic expression of lamprey klf17 in Xenopus embryos phenocopies Xenopus klf17. These data suggest that klf17 may have been the ancestral Klf factor that functioned in these gene regulatory networks in stem vertebrates.
Collapse
Affiliation(s)
- Sara Rigney
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Joshua R. York
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Carole LaBonne
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
- NSF-Simons National Institute for Theory and Mathematics in Biology, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
2
|
Jatzlau J, Do SN, Mees RA, Mendez PL, Khan RJ, Maas L, Ruiz L, Martin-Malpartida P, Macias MJ, Knaus P. Rare but specific: 5-bp composite motifs define SMAD binding in BMP signaling. BMC Biol 2025; 23:79. [PMID: 40082964 PMCID: PMC11907993 DOI: 10.1186/s12915-025-02183-1] [Citation(s) in RCA: 1] [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: 07/16/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Receptor-activated SMADs trimerize with SMAD4 to regulate context-dependent target gene expression. However, the presence of a single SMAD1/5/8 binding motif in cis-regulatory elements alone does not trigger transcription in native contexts. We hypothesize that binding to composite motifs in which at least two SMAD binding sites are in close proximity would be enough to induce transcription as this scenario allows the simultaneous interaction of at least two SMAD proteins, thereby increasing specificity and affinity. RESULTS Using more than 65 distinct firefly luciferase constructs, we delineated the minimal requirements for BMP-induced gene activation. We propose a model in which two SMAD-MH1 domains bind a SMAD-composite motif in a back-to-back fashion with a 5-bp distance between the SMAD-motifs on opposing DNA strands. However screening of SMAD1-bound regions across a variety of cell types highlights that these composite motifs are extremely uncommon, explaining below 1% of SMAD1 binding events. CONCLUSIONS Deviations from these minimal requirements prevent transcription and underline the need for co-transcription factors to achieve gene activation.
Collapse
Affiliation(s)
- Jerome Jatzlau
- Institute of Chemistry and Biochemistry, Freie Universitaet Berlin, Berlin, Germany
| | - Sophie-Nhi Do
- Institute of Chemistry and Biochemistry, Freie Universitaet Berlin, Berlin, Germany
| | - Rebeca A Mees
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac, 10, Barcelona, 08028, Spain
| | - Paul-Lennard Mendez
- Institute of Chemistry and Biochemistry, Freie Universitaet Berlin, Berlin, Germany
| | - Rameez Jabeer Khan
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac, 10, Barcelona, 08028, Spain
| | - Lukas Maas
- Institute of Chemistry and Biochemistry, Freie Universitaet Berlin, Berlin, Germany
| | - Lidia Ruiz
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac, 10, Barcelona, 08028, Spain
| | - Pau Martin-Malpartida
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac, 10, Barcelona, 08028, Spain
| | - Maria J Macias
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac, 10, Barcelona, 08028, Spain.
- ICREA, Passeig Lluís Companys 23, Barcelona, 08010, Spain.
| | - Petra Knaus
- Institute of Chemistry and Biochemistry, Freie Universitaet Berlin, Berlin, Germany.
| |
Collapse
|
3
|
Hucko LN, da Cruz NFS, Kemp PS, Mendoza-Santiesteban CE, Berrocal AM. Bilateral Iris and Chorioretinal Colobomas in a Child With Suspected Lamb-Shaffer Syndrome. Ophthalmic Surg Lasers Imaging Retina 2025:1-5. [PMID: 40163633 DOI: 10.3928/23258160-20250228-02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Lamb-Shaffer syndrome (LSS) is a rare neurodevelopmental disorder caused by haploin-sufficiency of the SRY-related HMB box (SOX5) gene. Most cases result from de novo variants; however, some cases follow autosomal dominant inheritance patterns. Classic features of LSS include global developmental delay, dysmorphic facial features, musculoskeletal abnormalities, and cardiac and genitourinary defects. Ophthalmic manifestations of LSS such as strabismus and optic nerve abnormalities have been described in approximately 55% of patients with LSS. We report a case of a 22-month-old boy with a history of bilateral iris and chorioretinal colobomas, bilateral lens subluxation, and strabismus and a past medical history of developmental delay, syndactyly, and hearing loss. Genetic testing identified a missense variant of uncertain significance in exon 14 of the SOX5 gene, which is linked to LSS. An inherited retinal disorders panel revealed the patient was a carrier of a pathogenic variant of the PHYH gene and a variant of uncertain significance of the COL9A2 gene. Results of the inherited retinal disorders panel did not identify variants implicated with iris and chorioretinal colobomas; therefore, we postulate colobomas as an additional ophthalmic manifestation of LSS. This case serves to broaden characterization of the LSS phenotype, while presenting an additional variant potentially associated with LSS. There is currently no treatment for LSS; however, robust understanding of LSS's heterogenous phenotypic and genetic profile will lead to improved knowledge of the syndrome and diagnostic aptitude. [Ophthalmic Surg Lasers Imaging Retina 2025;56:XX-XX.].
Collapse
|
4
|
Rigney S, York JR, LaBonne C. Krüppel-like Factors Play Essential Roles in Regulating Pluripotency and the Formation of Neural Crest Stem Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632647. [PMID: 39868152 PMCID: PMC11761489 DOI: 10.1101/2025.01.13.632647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The evolutionary transition from simple chordate body plans to complex vertebrate body plans was driven by the acquisition of the neural crest, a stem cell population that retains broad, multi-germ layer developmental potential long after most embryonic cells have become lineage restricted. We have previously shown that neural crest cells share significant gene regulatory architecture with pluripotent blastula stem cells. Here we examine the roles that Krüppel-like Family (Klf) transcription factors play in these stem cell populations. Although Klf4 has established roles in regulating pluripotency in mammalian stem cells cultures, we find that in Xenopus it is klf2 that is highly expressed in pluripotent blastula stem cells. klf2 expression is down-regulated as cells transition to a neural crest state while a related klf factor, klf17, is significantly up regulated in response to neural crest induction. We used gain and loss of function studies to compare the activities of these closely related factors and found that they have both shared and distinct activities. Inhibition of either klf2 or klf17 activity led to significantly expanded expression of pluripotency, neural plate border and neural crest factors in neurula stage embryos, leading us to hypothesize that klf factors regulate the exit from pluripotency and proper establishment of the boundary of the neural crest domain. To gain further insights into the role of klf factors in the evolution of the neural crest, we examined their expression in the jawless vertebrate, Petromyzon marinus ( sea lamprey). We find that lamprey have a klf2/4 and a klf17 gene, but that only klf17 is expressed in blastula and neural crest stem cells. Moreover, ectopic expression of lamprey klf17 in Xenopus embryos phenocopies Xenopus klf17 activity. These data suggest that klf17, rather than klf4, may have been the ancestral klf factor that functioned in these GRNs in stem vertebrates.
Collapse
|
5
|
York JR, Rao A, Huber PB, Schock EN, Montequin A, Rigney S, LaBonne C. Shared features of blastula and neural crest stem cells evolved at the base of vertebrates. Nat Ecol Evol 2024; 8:1680-1692. [PMID: 39060477 PMCID: PMC11520720 DOI: 10.1038/s41559-024-02476-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/18/2024] [Indexed: 07/28/2024]
Abstract
The neural crest is a vertebrate-specific stem cell population that helped drive the origin and evolution of vertebrates. A distinguishing feature of these cells is their multi-germ layer potential, which has parallels to another stem cell population-pluripotent stem cells of the vertebrate blastula. Here, we investigate the evolutionary origins of neural crest potential by comparing neural crest and pluripotency gene regulatory networks of a jawed vertebrate, Xenopus, and a jawless vertebrate, lamprey. We reveal an ancient evolutionary origin of shared regulatory factors in these gene regulatory networks that dates to the last common ancestor of extant vertebrates. Focusing on the key pluripotency factor pou5, we show that a lamprey pou5 orthologue is expressed in animal pole cells but is absent from neural crest. Both lamprey and Xenopus pou5 promote neural crest formation, suggesting that pou5 activity was lost from the neural crest of jawless vertebrates or acquired along the jawed vertebrate stem. Finally, we provide evidence that pou5 acquired novel, neural crest-enhancing activity after evolving from an ancestral pou3-like clade. This work provides evidence that both the neural crest and blastula pluripotency networks arose at the base of the vertebrates and that this may be linked to functional evolution of pou5.
Collapse
Affiliation(s)
- Joshua R York
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Anjali Rao
- Research Department, Gilead Sciences, Foster City, CA, USA
| | - Paul B Huber
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Elizabeth N Schock
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Andrew Montequin
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Sara Rigney
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Carole LaBonne
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA.
- National Institute for Theory and Mathematics in Biology, Chicago, IL, USA.
| |
Collapse
|
6
|
Huang J, Ren Q, Jiao L, Niu S, Liu C, Zhou J, Wu L, Yang Y. TMF suppresses chondrocyte hypertrophy in osteoarthritic cartilage by mediating the FOXO3a/BMPER pathway. Exp Ther Med 2024; 28:283. [PMID: 38800044 PMCID: PMC11117099 DOI: 10.3892/etm.2024.12571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Osteoarthritis (OA) is a disease of the joints, characterized by chronic inflammation, cartilage destruction and extracellular matrix (ECM) remodeling. Aberrant chondrocyte hypertrophy promotes cartilage destruction and OA development. Collagen X, the biomarker of chondrocyte hypertrophy, is upregulated by runt-related transcription factor 2 (Runx2), which is mediated by the bone morphogenetic protein 4 (BMP4)/Smad1 signaling pathway. BMP binding endothelial regulator (BMPER), a secreted glycoprotein, acts as an agonist of BMP4. 5,7,3',4'-tetramethoxyflavone (TMF) is a natural flavonoid derived from Murraya exotica L. Results of our previous study demonstrated that TMF exhibits chondroprotective effects against OA development through the activation of Forkhead box protein O3a (FOXO3a) expression. However, whether TMF suppresses chondrocyte hypertrophy through activation of FOXO3a expression and inhibition of BMPER/BMP4/Smad1 signaling remains unknown. Results of the present study revealed that TMF inhibited collagen X and Runx2 expression, inhibited BMPER/BMP4/Smad1 signaling, and activated FOXO3a expression; thus, protecting against chondrocyte hypertrophy and OA development. However, BMPER overexpression and FOXO3a knockdown impacted the protective effects of TMF. Thus, TMF inhibited chondrocyte hypertrophy in OA cartilage through mediating the FOXO3a/BMPER signaling pathway.
Collapse
Affiliation(s)
- Jishang Huang
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Qun Ren
- College of Pharmacy, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Linhui Jiao
- College of Pharmacy, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Shuo Niu
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Chenghong Liu
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Juan Zhou
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Longhuo Wu
- College of Pharmacy, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Yadong Yang
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| |
Collapse
|
7
|
Huber PB, Rao A, LaBonne C. BET activity plays an essential role in control of stem cell attributes in Xenopus. Development 2024; 151:dev202990. [PMID: 38884356 PMCID: PMC11266789 DOI: 10.1242/dev.202990] [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: 04/26/2024] [Accepted: 05/31/2024] [Indexed: 06/18/2024]
Abstract
Neural crest cells are a stem cell population unique to vertebrate embryos that retains broad multi-germ layer developmental potential through neurulation. Much remains to be learned about the genetic and epigenetic mechanisms that control the potency of neural crest cells. Here, we examine the role that epigenetic readers of the BET (bromodomain and extra terminal) family play in controlling the potential of pluripotent blastula and neural crest cells. We find that inhibiting BET activity leads to loss of pluripotency at blastula stages and a loss of neural crest at neurula stages. We compare the effects of HDAC (an eraser of acetylation marks) and BET (a reader of acetylation) inhibition and find that they lead to similar cellular outcomes through distinct effects on the transcriptome. Interestingly, loss of BET activity in cells undergoing lineage restriction is coupled to increased expression of genes linked to pluripotency and prolongs the competence of initially pluripotent cells to transit to a neural progenitor state. Together these findings advance our understanding of the epigenetic control of pluripotency and the formation of the vertebrate neural crest.
Collapse
Affiliation(s)
- Paul B. Huber
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
- National Institute for Theory and Mathematics in Biology, Northwestern University, Evanston, IL 60208, USA
| | - Anjali Rao
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Carole LaBonne
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
- National Institute for Theory and Mathematics in Biology, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
8
|
Humphreys PEA, Woods S, Bates N, Rooney KM, Mancini FE, Barclay C, O'Flaherty J, Martial FP, Domingos MAN, Kimber SJ. Optogenetic manipulation of BMP signaling to drive chondrogenic differentiation of hPSCs. Cell Rep 2023; 42:113502. [PMID: 38032796 DOI: 10.1016/j.celrep.2023.113502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/23/2023] [Accepted: 11/13/2023] [Indexed: 12/02/2023] Open
Abstract
Optogenetics is a rapidly advancing technology combining photochemical, optical, and synthetic biology to control cellular behavior. Together, sensitive light-responsive optogenetic tools and human pluripotent stem cell differentiation models have the potential to fine-tune differentiation and unpick the processes by which cell specification and tissue patterning are controlled by morphogens. We used an optogenetic bone morphogenetic protein (BMP) signaling system (optoBMP) to drive chondrogenic differentiation of human embryonic stem cells (hESCs). We engineered light-sensitive hESCs through CRISPR-Cas9-mediated integration of the optoBMP system into the AAVS1 locus. The activation of optoBMP with blue light, in lieu of BMP growth factors, resulted in the activation of BMP signaling mechanisms and upregulation of a chondrogenic phenotype, with significant transcriptional differences compared to cells in the dark. Furthermore, cells differentiated with light could form chondrogenic pellets consisting of a hyaline-like cartilaginous matrix. Our findings indicate the applicability of optogenetics for understanding human development and tissue engineering.
Collapse
Affiliation(s)
- Paul E A Humphreys
- Division of Cell Matrix & Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Steven Woods
- Division of Cell Matrix & Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Nicola Bates
- Division of Cell Matrix & Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Kirsty M Rooney
- Division of Cell Matrix & Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Fabrizio E Mancini
- Division of Cell Matrix & Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK; Department of Mechanical, Aerospace, and Civil Engineering, Faculty of Science and Engineering, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Cerys Barclay
- Division of Cell Matrix & Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Julieta O'Flaherty
- Division of Cell Matrix & Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Franck P Martial
- Division of Neuroscience & Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Marco A N Domingos
- Department of Mechanical, Aerospace, and Civil Engineering, Faculty of Science and Engineering, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Susan J Kimber
- Division of Cell Matrix & Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK.
| |
Collapse
|
9
|
York JR, Rao A, Huber PB, Schock EN, Montequin A, Rigney S, LaBonne C. Shared features of blastula and neural crest stem cells evolved at the base of vertebrates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572714. [PMID: 38187687 PMCID: PMC10769357 DOI: 10.1101/2023.12.21.572714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The neural crest is vertebrate-specific stem cell population that helped drive the origin and evolution of the vertebrate clade. A distinguishing feature of these stem cells is their multi-germ layer potential, which has drawn developmental and evolutionary parallels to another stem cell population-pluripotent embryonic stem cells (animal pole cells or ES cells) of the vertebrate blastula. Here, we investigate the evolutionary origins of neural crest potential by comparing neural crest and pluripotency gene regulatory networks (GRNs) in both jawed ( Xenopus ) and jawless (lamprey) vertebrates. Through comparative gene expression analysis and transcriptomics, we reveal an ancient evolutionary origin of shared regulatory factors between neural crest and pluripotency GRNs that dates back to the last common ancestor of extant vertebrates. Focusing on the key pluripotency factor pou5 (formerly oct4), we show that the lamprey genome encodes a pou5 ortholog that is expressed in animal pole cells, as in jawed vertebrates, but is absent from the neural crest. However, gain-of-function experiments show that both lamprey and Xenopus pou5 enhance neural crest formation, suggesting that pou5 was lost from the neural crest of jawless vertebrates. Finally, we show that pou5 is required for neural crest specification in jawed vertebrates and that it acquired novel neural crest-enhancing activity after evolving from an ancestral pou3 -like clade that lacks this functionality. We propose that a pluripotency-neural crest GRN was assembled in stem vertebrates and that the multi-germ layer potential of the neural crest evolved by deploying this regulatory program.
Collapse
|
10
|
Jatzlau J, Mendez PL, Altay A, Raaz L, Zhang Y, Mähr S, Sesver A, Reichenbach M, Mundlos S, Vingron M, Knaus P. Fluid shear stress-modulated chromatin accessibility reveals the mechano-dependency of endothelial SMAD1/5-mediated gene transcription. iScience 2023; 26:107405. [PMID: 37680470 PMCID: PMC10481294 DOI: 10.1016/j.isci.2023.107405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 04/20/2023] [Accepted: 07/12/2023] [Indexed: 09/09/2023] Open
Abstract
Bone morphogenetic protein (BMP) signaling and fluid shear stress (FSS) mediate complementary functions in vascular homeostasis and disease development. It remains to be shown whether altered chromatin accessibility downstream of BMP and FSS offers a crosstalk level to explain changes in SMAD-dependent transcription. Here, we employed ATAC-seq to analyze arterial endothelial cells stimulated with BMP9 and/or FSS. We found that BMP9-sensitive regions harbor non-palindromic GC-rich SMAD-binding elements (GGCTCC) and 69.7% of these regions become BMP-insensitive in the presence of FSS. While GATA and KLF transcription factor (TF) motifs are unique to BMP9- and FSS-sensitive regions, respectively, SOX motifs are common to both. Finally, we show that both SOX(13/18) and GATA(2/3/6) family members are directly upregulated by SMAD1/5. These findings highlight the mechano-dependency of SMAD-signaling by a sequential mechanism of first elevated pioneer TF expression, allowing subsequent chromatin opening to eventually providing accessibility to novel SMAD binding sites.
Collapse
Affiliation(s)
- Jerome Jatzlau
- Institute of Chemistry and Biochemistry - Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies (BSRT), 13353 Berlin, Germany
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Paul-Lennard Mendez
- Institute of Chemistry and Biochemistry - Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
- International Max-Planck Research School for Biology AND Computation (IMPRS-BAC), 14195 Berlin, Germany
| | - Aybuge Altay
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Lion Raaz
- Institute of Chemistry and Biochemistry - Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
- International Max-Planck Research School for Biology AND Computation (IMPRS-BAC), 14195 Berlin, Germany
- Institute of Medical and Human Genetics, Charité Universitätsmedizin, 13353 Berlin, Germany
| | - Yufei Zhang
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Sophia Mähr
- Institute of Chemistry and Biochemistry - Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Akin Sesver
- Institute of Chemistry and Biochemistry - Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Maria Reichenbach
- Institute of Chemistry and Biochemistry - Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Stefan Mundlos
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
- International Max-Planck Research School for Biology AND Computation (IMPRS-BAC), 14195 Berlin, Germany
- Institute of Medical and Human Genetics, Charité Universitätsmedizin, 13353 Berlin, Germany
| | - Martin Vingron
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
- International Max-Planck Research School for Biology AND Computation (IMPRS-BAC), 14195 Berlin, Germany
| | - Petra Knaus
- Institute of Chemistry and Biochemistry - Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies (BSRT), 13353 Berlin, Germany
- International Max-Planck Research School for Biology AND Computation (IMPRS-BAC), 14195 Berlin, Germany
| |
Collapse
|
11
|
Shigley C, Trivedi J, Meghani O, Owens BD, Jayasuriya CT. Suppressing Chondrocyte Hypertrophy to Build Better Cartilage. Bioengineering (Basel) 2023; 10:741. [PMID: 37370672 DOI: 10.3390/bioengineering10060741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Current clinical strategies for restoring cartilage defects do not adequately consider taking the necessary steps to prevent the formation of hypertrophic tissue at injury sites. Chondrocyte hypertrophy inevitably causes both macroscopic and microscopic level changes in cartilage, resulting in adverse long-term outcomes following attempted restoration. Repairing/restoring articular cartilage while minimizing the risk of hypertrophic neo tissue formation represents an unmet clinical challenge. Previous investigations have extensively identified and characterized the biological mechanisms that regulate cartilage hypertrophy with preclinical studies now beginning to leverage this knowledge to help build better cartilage. In this comprehensive article, we will provide a summary of these biological mechanisms and systematically review the most cutting-edge strategies for circumventing this pathological hallmark of osteoarthritis.
Collapse
Affiliation(s)
- Christian Shigley
- The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Jay Trivedi
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Ozair Meghani
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Brett D Owens
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
- Division of Sports Surgery, Department of Orthopaedic Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Chathuraka T Jayasuriya
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| |
Collapse
|
12
|
Patel I, Parchem RJ. Regulation of Oct4 in stem cells and neural crest cells. Birth Defects Res 2022; 114:983-1002. [PMID: 35365980 PMCID: PMC9525453 DOI: 10.1002/bdr2.2007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/25/2022] [Accepted: 03/14/2022] [Indexed: 12/30/2022]
Abstract
During embryonic development, cells gradually restrict their developmental potential as they exit pluripotency and differentiate into various cell types. The POU transcription factor Oct4 (encoded by Pou5f1) lies at the center of the pluripotency machinery that regulates stemness and differentiation in stem cells, and is required for reprogramming of somatic cells into induced pluripotent stem cells (iPSCs). Several studies have revealed that Oct4 and other stemness genes are also expressed in multipotent cell populations such as neural crest cells (NCCs), and are required to expand the NCC developmental potential. Transcriptional regulation of Oct4 has been studied extensively in stem cells during early embryonic development and reprogramming, but not in NCCs. Here, we review how Oct4 is regulated in pluripotent stem cells, and address some of the gaps in knowledge about regulation of the pluripotency network in NCCs.
Collapse
Affiliation(s)
- Ivanshi Patel
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTexasUSA,Stem Cells and Regenerative Medicine Center, Center for Cell and Gene TherapyBaylor College of MedicineHoustonTexasUSA,Dan L Duncan Comprehensive Cancer CenterBaylor College of MedicineHoustonTexasUSA
| | - Ronald J. Parchem
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTexasUSA,Stem Cells and Regenerative Medicine Center, Center for Cell and Gene TherapyBaylor College of MedicineHoustonTexasUSA,Dan L Duncan Comprehensive Cancer CenterBaylor College of MedicineHoustonTexasUSA
| |
Collapse
|
13
|
Ehnes DD, Alghadeer A, Hanson-Drury S, Zhao YT, Tilmes G, Mathieu J, Ruohola-Baker H. Sci-Seq of Human Fetal Salivary Tissue Introduces Human Transcriptional Paradigms and a Novel Cell Population. FRONTIERS IN DENTAL MEDICINE 2022; 3:887057. [PMID: 36540608 PMCID: PMC9762771 DOI: 10.3389/fdmed.2022.887057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023] Open
Abstract
Multiple pathologies and non-pathological factors can disrupt the function of the non-regenerative human salivary gland including cancer and cancer therapeutics, autoimmune diseases, infections, pharmaceutical side effects, and traumatic injury. Despite the wide range of pathologies, no therapeutic or regenerative approaches exist to address salivary gland loss, likely due to significant gaps in our understanding of salivary gland development. Moreover, identifying the tissue of origin when diagnosing salivary carcinomas requires an understanding of human fetal development. Using computational tools, we identify developmental branchpoints, a novel stem cell-like population, and key signaling pathways in the human developing salivary glands by analyzing our human fetal single-cell sequencing data. Trajectory and transcriptional analysis suggest that the earliest progenitors yield excretory duct and myoepithelial cells and a transitional population that will yield later ductal cell types. Importantly, this single-cell analysis revealed a previously undescribed population of stem cell-like cells that are derived from SD and expresses high levels of genes associated with stem cell-like function. We have observed these rare cells, not in a single niche location but dispersed within the developing duct at later developmental stages. Our studies introduce new human-specific developmental paradigms for the salivary gland and lay the groundwork for the development of translational human therapeutics.
Collapse
Affiliation(s)
- Devon Duron Ehnes
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cells and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
| | - Ammar Alghadeer
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cells and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
- Department of Biomedical Dental Sciences, College of Dentistry, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Sesha Hanson-Drury
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cells and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
- Department of Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, United States
| | - Yan Ting Zhao
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cells and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
- Department of Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, United States
| | - Gwen Tilmes
- Institute for Stem Cells and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
| | - Julie Mathieu
- Institute for Stem Cells and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
- Department of Comparative Medicine, University of Washington, Seattle, WA, United States
| | - Hannele Ruohola-Baker
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cells and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
- Department of Biomedical Dental Sciences, College of Dentistry, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| |
Collapse
|
14
|
Schock EN, LaBonne C. Sorting Sox: Diverse Roles for Sox Transcription Factors During Neural Crest and Craniofacial Development. Front Physiol 2020; 11:606889. [PMID: 33424631 PMCID: PMC7793875 DOI: 10.3389/fphys.2020.606889] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/09/2020] [Indexed: 12/31/2022] Open
Abstract
Sox transcription factors play many diverse roles during development, including regulating stem cell states, directing differentiation, and influencing the local chromatin landscape. Of the twenty vertebrate Sox factors, several play critical roles in the development the neural crest, a key vertebrate innovation, and the subsequent formation of neural crest-derived structures, including the craniofacial complex. Herein, we review the specific roles for individual Sox factors during neural crest cell formation and discuss how some factors may have been essential for the evolution of the neural crest. Additionally, we describe how Sox factors direct neural crest cell differentiation into diverse lineages such as melanocytes, glia, and cartilage and detail their involvement in the development of specific craniofacial structures. Finally, we highlight several SOXopathies associated with craniofacial phenotypes.
Collapse
Affiliation(s)
- Elizabeth N. Schock
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, United States
| | - Carole LaBonne
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, United States
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL, United States
| |
Collapse
|
15
|
Perera SN, Kerosuo L. On the road again: Establishment and maintenance of stemness in the neural crest from embryo to adulthood. STEM CELLS (DAYTON, OHIO) 2020; 39:7-25. [PMID: 33017496 PMCID: PMC7821161 DOI: 10.1002/stem.3283] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/08/2020] [Accepted: 09/11/2020] [Indexed: 12/22/2022]
Abstract
Unique to vertebrates, the neural crest (NC) is an embryonic stem cell population that contributes to a greatly expanding list of derivatives ranging from neurons and glia of the peripheral nervous system, facial cartilage and bone, pigment cells of the skin to secretory cells of the endocrine system. Here, we focus on what is specifically known about establishment and maintenance of NC stemness and ultimate fate commitment mechanisms, which could help explain its exceptionally high stem cell potential that exceeds the "rules set during gastrulation." In fact, recent discoveries have shed light on the existence of NC cells that coexpress commonly accepted pluripotency factors like Nanog, Oct4/PouV, and Klf4. The coexpression of pluripotency factors together with the exceptional array of diverse NC derivatives encouraged us to propose a new term "pleistopotent" (Greek for abundant, a substantial amount) to be used to reflect the uniqueness of the NC as compared to other post-gastrulation stem cell populations in the vertebrate body, and to differentiate them from multipotent lineage restricted stem cells. We also discuss studies related to the maintenance of NC stemness within the challenging context of being a transient and thus a constantly changing population of stem cells without a permanent niche. The discovery of the stem cell potential of Schwann cell precursors as well as multiple adult NC-derived stem cell reservoirs during the past decade has greatly increased our understanding of how NC cells contribute to tissues formed after its initial migration stage in young embryos.
Collapse
Affiliation(s)
- Surangi N Perera
- Neural Crest Development and Disease Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Laura Kerosuo
- Neural Crest Development and Disease Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
16
|
Knock-down Sox5 suppresses porcine adipogenesis through BMP R-Smads signal pathway. Biochem Biophys Res Commun 2020; 527:574-580. [PMID: 32423805 DOI: 10.1016/j.bbrc.2020.04.125] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 04/23/2020] [Indexed: 12/27/2022]
|
17
|
Prasad MS, Charney RM, García-Castro MI. Specification and formation of the neural crest: Perspectives on lineage segregation. Genesis 2019; 57:e23276. [PMID: 30576078 PMCID: PMC6570420 DOI: 10.1002/dvg.23276] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 12/21/2022]
Abstract
The neural crest is a fascinating embryonic population unique to vertebrates that is endowed with remarkable differentiation capacity. Thought to originate from ectodermal tissue, neural crest cells generate neurons and glia of the peripheral nervous system, and melanocytes throughout the body. However, the neural crest also generates many ectomesenchymal derivatives in the cranial region, including cell types considered to be of mesodermal origin such as cartilage, bone, and adipose tissue. These ectomesenchymal derivatives play a critical role in the formation of the vertebrate head, and are thought to be a key attribute at the center of vertebrate evolution and diversity. Further, aberrant neural crest cell development and differentiation is the root cause of many human pathologies, including cancers, rare syndromes, and birth malformations. In this review, we discuss the current findings of neural crest cell ontogeny, and consider tissue, cell, and molecular contributions toward neural crest formation. We further provide current perspectives into the molecular network involved during the segregation of the neural crest lineage.
Collapse
Affiliation(s)
- Maneeshi S Prasad
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California
| | - Rebekah M Charney
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California
| | - Martín I García-Castro
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California
| |
Collapse
|
18
|
Watanabe T, Yamamoto T, Tsukano K, Hirano S, Horikawa A, Michiue T. Fam46a regulates BMP-dependent pre-placodal ectoderm differentiation in Xenopus. Development 2018; 145:dev.166710. [PMID: 30291163 DOI: 10.1242/dev.166710] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 09/06/2018] [Indexed: 12/30/2022]
Abstract
The pre-placodal ectoderm (PPE) is a specialized ectodermal region which gives rise to the sensory organs and other systems. The PPE is induced from the neural plate border during neurulation, but the molecular mechanism of PPE formation is not fully understood. In this study, we examined the role of a newly identified PPE gene, Fam46a, during embryogenesis. Fam46a contains a nucleoside triphosphate transferase domain, but its function in early development was previously unclear. We show that Fam46a is expressed in the PPE in Xenopus embryos, and Fam46a knockdown induces abnormalities in the eye formation and the body color. At the neurula stage, Fam46a upregulates the expression of PPE genes and inhibits neural crest formation. We also show that Fam46a physically interacts with Smad1/Smad4 and positively regulates BMP signaling. From these results, we conclude that Fam46a is required for PPE formation via the positive regulation of BMP signaling. Our study provides a new mechanism of ectodermal patterning via cell-autonomous regulation of BMP signaling in the PPE.
Collapse
Affiliation(s)
- Tomoko Watanabe
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Takayoshi Yamamoto
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Kohei Tsukano
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Sayuki Hirano
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Ayumi Horikawa
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Tatsuo Michiue
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| |
Collapse
|
19
|
Rogers CD, Nie S. Specifying neural crest cells: From chromatin to morphogens and factors in between. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2018; 7:e322. [PMID: 29722151 PMCID: PMC6215528 DOI: 10.1002/wdev.322] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 03/26/2018] [Accepted: 03/27/2018] [Indexed: 12/16/2022]
Abstract
Neural crest (NC) cells are a stem-like multipotent population of progenitor cells that are present in vertebrate embryos, traveling to various regions in the developing organism. Known as the "fourth germ layer," these cells originate in the ectoderm between the neural plate (NP), which will become the brain and spinal cord, and nonneural tissues that will become the skin and the sensory organs. NC cells can differentiate into more than 30 different derivatives in response to the appropriate signals including, but not limited to, craniofacial bone and cartilage, sensory nerves and ganglia, pigment cells, and connective tissue. The molecular and cellular mechanisms that control the induction and specification of NC cells include epigenetic control, multiple interactive and redundant transcriptional pathways, secreted signaling molecules, and adhesion molecules. NC cells are important not only because they transform into a wide variety of tissue types, but also because their ability to detach from their epithelial neighbors and migrate throughout developing embryos utilizes mechanisms similar to those used by metastatic cancer cells. In this review, we discuss the mechanisms required for the induction and specification of NC cells in various vertebrate species, focusing on the roles of early morphogenesis, cell adhesion, signaling from adjacent tissues, and the massive transcriptional network that controls the formation of these amazing cells. This article is categorized under: Nervous System Development > Vertebrates: General Principles Gene Expression and Transcriptional Hierarchies > Regulatory Mechanisms Gene Expression and Transcriptional Hierarchies > Gene Networks and Genomics Signaling Pathways > Cell Fate Signaling.
Collapse
Affiliation(s)
- Crystal D. Rogers
- Department of Biology, College of Science and Mathematics, California State University Northridge, Northridge, California
| | - Shuyi Nie
- School of Biological Sciences and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
20
|
Buitrago-Delgado E, Schock EN, Nordin K, LaBonne C. A transition from SoxB1 to SoxE transcription factors is essential for progression from pluripotent blastula cells to neural crest cells. Dev Biol 2018; 444:50-61. [PMID: 30144418 DOI: 10.1016/j.ydbio.2018.08.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 08/10/2018] [Accepted: 08/21/2018] [Indexed: 01/30/2023]
Abstract
The neural crest is a stem cell population unique to vertebrate embryos that gives rise to derivatives from multiple embryonic germ layers. The molecular underpinnings of potency that govern neural crest potential are highly conserved with that of pluripotent blastula stem cells, suggesting that neural crest cells may have evolved through retention of aspects of the pluripotency gene regulatory network (GRN). A striking difference in the regulatory factors utilized in pluripotent blastula cells and neural crest cells is the deployment of different sub-families of Sox transcription factors; SoxB1 factors play central roles in the pluripotency of naïve blastula and ES cells, whereas neural crest cells require SoxE function. Here we explore the shared and distinct activities of these factors to shed light on the role that this molecular hand-off of Sox factor activity plays in the genesis of neural crest and the lineages derived from it. Our findings provide evidence that SoxB1 and SoxE factors have both overlapping and distinct activities in regulating pluripotency and lineage restriction in the embryo. We hypothesize that SoxE factors may transiently replace SoxB1 factors to control pluripotency in neural crest cells, and then poise these cells to contribute to glial, chondrogenic and melanocyte lineages at stages when SoxB1 factors promote neuronal progenitor formation.
Collapse
Affiliation(s)
- Elsy Buitrago-Delgado
- Dept. of Molecular Biosciences, Northwestern University, Evanston, IL 60208, United States
| | - Elizabeth N Schock
- Dept. of Molecular Biosciences, Northwestern University, Evanston, IL 60208, United States
| | - Kara Nordin
- Dept. of Molecular Biosciences, Northwestern University, Evanston, IL 60208, United States
| | - Carole LaBonne
- Dept. of Molecular Biosciences, Northwestern University, Evanston, IL 60208, United States; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Evanston, IL 60208, United States.
| |
Collapse
|
21
|
Rao A, LaBonne C. Histone deacetylase activity has an essential role in establishing and maintaining the vertebrate neural crest. Development 2018; 145:dev.163386. [PMID: 30002130 DOI: 10.1242/dev.163386] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 07/04/2018] [Indexed: 12/24/2022]
Abstract
The neural crest, a progenitor population that drove vertebrate evolution, retains the broad developmental potential of the blastula cells it is derived from, even as neighboring cells undergo lineage restriction. The mechanisms that enable these cells to preserve their developmental potential remain poorly understood. Here, we explore the role of histone deacetylase (HDAC) activity in this process in Xenopus We show that HDAC activity is essential for the formation of neural crest, as well as for proper patterning of the early ectoderm. The requirement for HDAC activity initiates in naïve blastula cells; HDAC inhibition causes loss of pluripotency gene expression and blocks the ability of blastula stem cells to contribute to lineages of the three embryonic germ layers. We find that pluripotent naïve blastula cells and neural crest cells are both characterized by low levels of histone acetylation, and show that increasing HDAC1 levels enhance the ability of blastula cells to be reprogrammed to a neural crest state. Together, these findings elucidate a previously uncharacterized role for HDAC activity in establishing the neural crest stem cell state.
Collapse
Affiliation(s)
- Anjali Rao
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Carole LaBonne
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA .,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
22
|
Rogers CD, Sorrells LK, Bronner ME. A catenin-dependent balance between N-cadherin and E-cadherin controls neuroectodermal cell fate choices. Mech Dev 2018; 152:44-56. [PMID: 30009960 PMCID: PMC6112866 DOI: 10.1016/j.mod.2018.07.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/16/2018] [Accepted: 07/02/2018] [Indexed: 01/19/2023]
Abstract
Characterizing endogenous protein expression, interaction and function, this study identifies in vivo interactions and competitive balance between N-cadherin and E-cadherin in developing avian (Gallus gallus) neural and neural crest cells. Numerous cadherin proteins, including neural cadherin (Ncad) and epithelial cadherin (Ecad), are expressed in the developing neural plate as well as in neural crest cells as they delaminate from the newly closed neural tube. To clarify independent or coordinate function during development, we examined their expression in the cranial region. The results revealed surprising overlap and distinct localization of Ecad and Ncad in the neural tube. Using a proximity ligation assay and co-immunoprecipitation, we found that Ncad and Ecad formed heterotypic complexes in the developing neural tube, and that modulation of Ncad levels led to reciprocal gain or reduction of Ecad protein, which then alters ectodermal cell fate. Here, we demonstrate that the balance of Ecad and Ncad is dependent upon the availability of β-catenin proteins, and that alteration of either classical cadherin modifies the proportions of the neural crest and neuroectodermal cells that are specified.
Collapse
Affiliation(s)
- Crystal D Rogers
- Department of Biology, California State University, Northridge, Northridge, CA 91330, United States of America.
| | - Lisa K Sorrells
- Department of Biology, California State University, Northridge, Northridge, CA 91330, United States of America.
| | - Marianne E Bronner
- Division of Biology and Biological Engineering 139-74, California Institute of Technology, Pasadena, CA 91125, United States of America.
| |
Collapse
|
23
|
Xenopus SOX5 enhances myogenic transcription indirectly through transrepression. Dev Biol 2018; 442:262-275. [PMID: 30071218 DOI: 10.1016/j.ydbio.2018.07.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/16/2018] [Accepted: 07/28/2018] [Indexed: 02/06/2023]
Abstract
In anamniotes, somite compartimentalization in the lateral somitic domain leads simultaneously to myotome and dermomyotome formation. In the myotome, Xenopus Sox5 is co-expressed with Myod1 in the course of myogenic differentiation. Here, we studied the function of Sox5 using a Myod1-induced myogenic transcription assay in pluripotent cells of animal caps. We found that Sox5 enhances myogenic transcription of muscle markers Des, Actc1, Ckm and MyhE3. The use of chimeric transactivating or transrepressive Sox5 proteins indicates that Sox5 acts as a transrepressor and indirectly stimulates myogenic transcription except for the slow muscle-specific genes Myh7L, Myh7S, Myl2 and Tnnc1. We showed that this role is shared by Sox6, which is structurally similar to Sox5, both belonging to the SoxD subfamily of transcription factors. Moreover, Sox5 can antagonize the inhibitory function of Meox2 on myogenic differentiation. Meox2 which is a dermomyotome marker, represses myogenic transcription in Myod-induced myogenic transcription assay and in Nodal5-induced mesoderm from animal cap assay. The inhibitory function of Meox2 and the pro-myogenic function of Sox5 were confirmed during Xenopus normal development by the use of translation-blocking oligomorpholinos and dexamethasone inducible chimeric Sox5 and Meox2 proteins. We have therefore identified a new function for SoxD proteins in muscle cells, which can indirectly enhance myogenic transcription through transrepression, in addition to the previously identified function as a direct repressor of slow muscle-specific genes.
Collapse
|
24
|
Castro Colabianchi AM, Revinski DR, Encinas PI, Baez MV, Monti RJ, Rodríguez Abinal M, Kodjabachian L, Franchini LF, López SL. Notch1 is asymmetrically distributed from the beginning of embryogenesis and controls the ventral center. Development 2018; 145:dev.159368. [PMID: 29866901 DOI: 10.1242/dev.159368] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 05/25/2018] [Indexed: 12/14/2022]
Abstract
Based on functional evidence, we have previously demonstrated that early ventral Notch1 activity restricts dorsoanterior development in Xenopus We found that Notch1 has ventralizing properties and abolishes the dorsalizing activity of β-catenin by reducing its steady state levels, in a process that does not require β-catenin phosphorylation by glycogen synthase kinase 3β. In the present work, we demonstrate that Notch1 mRNA and protein are enriched in the ventral region from the beginning of embryogenesis in Xenopus This is the earliest sign of ventral development, preceding the localized expression of wnt8a, bmp4 and Ventx genes in the ventral center and the dorsal accumulation of nuclear β-catenin. Knockdown experiments indicate that Notch1 is necessary for the normal expression of genes essential for ventral-posterior development. These results indicate that during early embryogenesis ventrally located Notch1 promotes the development of the ventral center. Together with our previous evidence, these results suggest that ventral enrichment of Notch1 underlies the process by which Notch1 participates in restricting nuclear accumulation of β-catenin to the dorsal side.
Collapse
Affiliation(s)
- Aitana M Castro Colabianchi
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Biología Celular y Neurociencias 'Prof. E. De Robertis' (IBCN), Facultad de Medicina. Laboratorio de Embriología Molecular 'Prof. Dr. Andrés E. Carrasco', C1121ABG Buenos Aires, Argentina
| | - Diego R Revinski
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Biología Celular y Neurociencias 'Prof. E. De Robertis' (IBCN), Facultad de Medicina. Laboratorio de Embriología Molecular 'Prof. Dr. Andrés E. Carrasco', C1121ABG Buenos Aires, Argentina.,Aix Marseille Université, CNRS, IBDM, 13288 Marseille, France
| | - Paula I Encinas
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Biología Celular y Neurociencias 'Prof. E. De Robertis' (IBCN), Facultad de Medicina. Laboratorio de Embriología Molecular 'Prof. Dr. Andrés E. Carrasco', C1121ABG Buenos Aires, Argentina
| | - María Verónica Baez
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Biología Celular y Neurociencias 'Prof. E. De Robertis' (IBCN), Facultad de Medicina. Laboratorio de Embriología Molecular 'Prof. Dr. Andrés E. Carrasco', C1121ABG Buenos Aires, Argentina
| | - Renato J Monti
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Biología Celular y Neurociencias 'Prof. E. De Robertis' (IBCN), Facultad de Medicina. Laboratorio de Embriología Molecular 'Prof. Dr. Andrés E. Carrasco', C1121ABG Buenos Aires, Argentina
| | - Mateo Rodríguez Abinal
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Biología Celular y Neurociencias 'Prof. E. De Robertis' (IBCN), Facultad de Medicina. Laboratorio de Embriología Molecular 'Prof. Dr. Andrés E. Carrasco', C1121ABG Buenos Aires, Argentina
| | | | - Lucía F Franchini
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428ADN Buenos Aires, Argentina
| | - Silvia L López
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Biología Celular y Neurociencias 'Prof. E. De Robertis' (IBCN), Facultad de Medicina. Laboratorio de Embriología Molecular 'Prof. Dr. Andrés E. Carrasco', C1121ABG Buenos Aires, Argentina
| |
Collapse
|
25
|
Geary L, LaBonne C. FGF mediated MAPK and PI3K/Akt Signals make distinct contributions to pluripotency and the establishment of Neural Crest. eLife 2018; 7:33845. [PMID: 29350613 PMCID: PMC5790379 DOI: 10.7554/elife.33845] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 01/15/2018] [Indexed: 12/12/2022] Open
Abstract
Early vertebrate embryos possess cells with the potential to generate all embryonic cell types. While this pluripotency is progressively lost as cells become lineage restricted, Neural Crest cells retain broad developmental potential. Here, we provide novel insights into signals essential for both pluripotency and neural crest formation in Xenopus. We show that FGF signaling controls a subset of genes expressed by pluripotent blastula cells, and find a striking switch in the signaling cascades activated by FGF signaling as cells lose pluripotency and commence lineage restriction. Pluripotent cells display and require Map Kinase signaling, whereas PI3 Kinase/Akt signals increase as developmental potential is restricted, and are required for transit to certain lineage restricted states. Importantly, retaining a high Map Kinase/low Akt signaling profile is essential for establishing Neural Crest stem cells. These findings shed important light on the signal-mediated control of pluripotency and the molecular mechanisms governing genesis of Neural Crest.
Collapse
Affiliation(s)
- Lauren Geary
- Department of Molecular Biosciences, Northwestern University, Evanston, United States
| | - Carole LaBonne
- Department of Molecular Biosciences, Northwestern University, Evanston, United States.,Robert H Lurie Comprehensive Cancer Center, Northwestern University, Evanston, United States
| |
Collapse
|
26
|
Kahata K, Dadras MS, Moustakas A. TGF-β Family Signaling in Epithelial Differentiation and Epithelial-Mesenchymal Transition. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a022194. [PMID: 28246184 DOI: 10.1101/cshperspect.a022194] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Epithelia exist in the animal body since the onset of embryonic development; they generate tissue barriers and specify organs and glands. Through epithelial-mesenchymal transitions (EMTs), epithelia generate mesenchymal cells that form new tissues and promote healing or disease manifestation when epithelial homeostasis is challenged physiologically or pathologically. Transforming growth factor-βs (TGF-βs), activins, bone morphogenetic proteins (BMPs), and growth and differentiation factors (GDFs) have been implicated in the regulation of epithelial differentiation. These TGF-β family ligands are expressed and secreted at sites where the epithelium interacts with the mesenchyme and provide paracrine queues from the mesenchyme to the neighboring epithelium, helping the specification of differentiated epithelial cell types within an organ. TGF-β ligands signal via Smads and cooperating kinase pathways and control the expression or activities of key transcription factors that promote either epithelial differentiation or mesenchymal transitions. In this review, we discuss evidence that illustrates how TGF-β family ligands contribute to epithelial differentiation and induce mesenchymal transitions, by focusing on the embryonic ectoderm and tissues that form the external mammalian body lining.
Collapse
Affiliation(s)
- Kaoru Kahata
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Mahsa Shahidi Dadras
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, SE-751 24 Uppsala, Sweden.,Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Aristidis Moustakas
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, SE-751 24 Uppsala, Sweden.,Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, SE-751 23 Uppsala, Sweden
| |
Collapse
|
27
|
Ampuja M, Kallioniemi A. Transcription factors-Intricate players of the bone morphogenetic protein signaling pathway. Genes Chromosomes Cancer 2017; 57:3-11. [DOI: 10.1002/gcc.22502] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/28/2017] [Accepted: 08/28/2017] [Indexed: 12/14/2022] Open
Affiliation(s)
- M. Ampuja
- BioMediTech Institute and Faculty of Medicine and Life Sciences; University of Tampere; Tampere Finland
| | - Anne Kallioniemi
- BioMediTech Institute and Faculty of Medicine and Life Sciences; University of Tampere; Tampere Finland
- Fimlab Laboratories; Tampere Finland
| |
Collapse
|
28
|
Budi EH, Duan D, Derynck R. Transforming Growth Factor-β Receptors and Smads: Regulatory Complexity and Functional Versatility. Trends Cell Biol 2017; 27:658-672. [PMID: 28552280 DOI: 10.1016/j.tcb.2017.04.005] [Citation(s) in RCA: 223] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 02/06/2023]
Abstract
Transforming growth factor (TGF)-β family proteins control cell physiology, proliferation, and growth, and direct cell differentiation, thus playing key roles in normal development and disease. The mechanisms of how TGF-β family ligands interact with heteromeric complexes of cell surface receptors to then activate Smad signaling that directs changes in gene expression are often seen as established. Even though TGF-β-induced Smad signaling may be seen as a linear signaling pathway with predictable outcomes, this pathway provides cells with a versatile means to induce different cellular responses. Fundamental questions remain as to how, at the molecular level, TGF-β and TGF-β family proteins activate the receptor complexes and induce a context-dependent diversity of cell responses. Among the areas of progress, we summarize new insights into how cells control TGF-β responsiveness by controlling the TGF-β receptors, and into the key roles and versatility of Smads in directing cell differentiation and cell fate selection.
Collapse
Affiliation(s)
- Erine H Budi
- Department of Cell and Tissue Biology, and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco CA 94143, USA
| | - Dana Duan
- Department of Cell and Tissue Biology, and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco CA 94143, USA
| | - Rik Derynck
- Department of Cell and Tissue Biology, and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco CA 94143, USA.
| |
Collapse
|
29
|
Li A, Hooli B, Mullin K, Tate RE, Bubnys A, Kirchner R, Chapman B, Hofmann O, Hide W, Tanzi RE. Silencing of the Drosophila ortholog of SOX5 leads to abnormal neuronal development and behavioral impairment. Hum Mol Genet 2017; 26:1472-1482. [PMID: 28186563 DOI: 10.1093/hmg/ddx051] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 02/07/2017] [Indexed: 01/27/2023] Open
Abstract
SOX5 encodes a transcription factor that is expressed in multiple tissues including heart, lung and brain. Mutations in SOX5 have been previously found in patients with amyotrophic lateral sclerosis (ALS) and developmental delay, intellectual disability and dysmorphic features. To characterize the neuronal role of SOX5, we silenced the Drosophila ortholog of SOX5, Sox102F, by RNAi in various neuronal subtypes in Drosophila. Silencing of Sox102F led to misorientated and disorganized michrochaetes, neurons with shorter dendritic arborization (DA) and reduced complexity, diminished larval peristaltic contractions, loss of neuromuscular junction bouton structures, impaired olfactory perception, and severe neurodegeneration in brain. Silencing of SOX5 in human SH-SY5Y neuroblastoma cells resulted in a significant repression of WNT signaling activity and altered expression of WNT-related genes. Genetic association and meta-analyses of the results in several large family-based and case-control late-onset familial Alzheimer's disease (LOAD) samples of SOX5 variants revealed several variants that show significant association with AD disease status. In addition, analysis for rare and highly penetrate functional variants revealed four novel variants/mutations in SOX5, which taken together with functional prediction analysis, suggests a strong role of SOX5 causing AD in the carrier families. Collectively, these findings indicate that SOX5 is a novel candidate gene for LOAD with an important role in neuronal function. The genetic findings warrant further studies to identify and characterize SOX5 variants that confer risk for AD, ALS and intellectual disability.
Collapse
Affiliation(s)
- Airong Li
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, MassGeneral Institute for Neurodegenerative Diseases, Charlestown, MA 02129, USA
| | - Basavaraj Hooli
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, MassGeneral Institute for Neurodegenerative Diseases, Charlestown, MA 02129, USA
| | - Kristina Mullin
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, MassGeneral Institute for Neurodegenerative Diseases, Charlestown, MA 02129, USA
| | - Rebecca E Tate
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, MassGeneral Institute for Neurodegenerative Diseases, Charlestown, MA 02129, USA
| | - Adele Bubnys
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, MassGeneral Institute for Neurodegenerative Diseases, Charlestown, MA 02129, USA
| | - Rory Kirchner
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Brad Chapman
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Oliver Hofmann
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA.,Center for Cancer Research, University of Melbourne, Melbourne 3000, Australia and
| | - Winston Hide
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA.,Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, MassGeneral Institute for Neurodegenerative Diseases, Charlestown, MA 02129, USA
| |
Collapse
|
30
|
Schille C, Schambony A. Signaling pathways and tissue interactions in neural plate border formation. NEUROGENESIS 2017; 4:e1292783. [PMID: 28352644 DOI: 10.1080/23262133.2017.1292783] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/02/2017] [Accepted: 02/02/2017] [Indexed: 02/04/2023]
Abstract
The neural crest is a transient cell population that gives rise to various cell types of multiple tissues and organs in the vertebrate embryo. Neural crest cells arise from the neural plate border, a region localized at the lateral borders of the prospective neural plate. Temporally and spatially coordinated interaction with the adjacent tissues, the non-neural ectoderm, the neural plate and the prospective dorsolateral mesoderm, is required for neural plate border specification. Signaling molecules, namely BMP, Wnt and FGF ligands and corresponding antagonists are derived from these tissues and interact to induce the expression of neural plate border specific genes. The present mini-review focuses on the current understanding of how the NPB territory is formed and accentuates the need for coordinated interaction of BMP and Wnt signaling pathways and precise tissue communication that are required for the definition of the prospective NC in the competent ectoderm.
Collapse
Affiliation(s)
- Carolin Schille
- Biology Department, Developmental Biology, Friedrich-Alexander University Erlangen-Nuremberg , Erlangen, Germany
| | - Alexandra Schambony
- Biology Department, Developmental Biology, Friedrich-Alexander University Erlangen-Nuremberg , Erlangen, Germany
| |
Collapse
|
31
|
Murko C, Bronner ME. Tissue specific regulation of the chick Sox10E1 enhancer by different Sox family members. Dev Biol 2016; 422:47-57. [PMID: 28012818 DOI: 10.1016/j.ydbio.2016.12.004] [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: 06/16/2016] [Revised: 12/02/2016] [Accepted: 12/03/2016] [Indexed: 01/15/2023]
Abstract
The transcription factor Sox10 is a key regulator of vertebrate neural crest development and serves crucial functions in the differentiation of multiple neural crest lineages. In the chick neural crest, two cis-regulatory elements have been identified that mediate Sox10 expression: Sox10E2, which initiates expression in cranial neural crest; Sox10E1 driving expression in vagal and trunk neural crest. Both also mediate Sox10 expression in the otic placode. Here, we have dissected and analyzed the Sox10E1 enhancer element to identify upstream regulatory inputs. Via mutational analysis, we found two critical Sox sites with differential impact on trunk versus otic Sox10E1 mediated reporter expression. Mutation of a combined SoxD/E motif was sufficient to completely abolish neural crest but not ear enhancer activity. However, mutation of both the SoxD/E and another SoxE site eliminated otic Sox10E1 expression. Loss-of-function experiments reveal Sox5 and Sox8 as critical inputs for trunk neural crest enhancer activity, but only Sox8 for its activity in the ear. Finally, we show by ChIP and co-immunoprecipitation that Sox5 directly binds to the SoxD/E site, and that it can interact with Sox8, further supporting their combinatorial role in activation of Sox10E1 in the trunk neural crest. The results reveal important tissue-specific inputs into Sox10 expression in the developing embryo.
Collapse
Affiliation(s)
- Christina Murko
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, United States
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, United States.
| |
Collapse
|
32
|
The Signaling Pathways Involved in Chondrocyte Differentiation and Hypertrophic Differentiation. Stem Cells Int 2016; 2016:2470351. [PMID: 28074096 PMCID: PMC5198191 DOI: 10.1155/2016/2470351] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 11/22/2016] [Indexed: 12/19/2022] Open
Abstract
Chondrocytes communicate with each other mainly via diffusible signals rather than direct cell-to-cell contact. The chondrogenic differentiation of mesenchymal stem cells (MSCs) is well regulated by the interactions of varieties of growth factors, cytokines, and signaling molecules. A number of critical signaling molecules have been identified to regulate the differentiation of chondrocyte from mesenchymal progenitor cells to their terminal maturation of hypertrophic chondrocytes, including bone morphogenetic proteins (BMPs), SRY-related high-mobility group-box gene 9 (Sox9), parathyroid hormone-related peptide (PTHrP), Indian hedgehog (Ihh), fibroblast growth factor receptor 3 (FGFR3), and β-catenin. Except for these molecules, other factors such as adenosine, O2 tension, and reactive oxygen species (ROS) also have a vital role in cartilage formation and chondrocyte maturation. Here, we outlined the complex transcriptional network and the function of key factors in this network that determine and regulate the genetic program of chondrogenesis and chondrocyte differentiation.
Collapse
|
33
|
Ji EH, Kim J. SoxD Transcription Factors: Multifaceted Players of Neural Development. Int J Stem Cells 2016; 9:3-8. [PMID: 27426080 PMCID: PMC4961098 DOI: 10.15283/ijsc.2016.9.1.3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2016] [Indexed: 01/05/2023] Open
Abstract
SoxD transcription factor subfamily includes three members, Sox5, Sox6, and Sox13. Like other Sox genes, they contain the High-Mobility-Group (HMG) box as the DNA binding domain but in addition feature the subgroup-specific leucine zipper motif. SoxD genes are expressed in diverse cell types in multiple organs during embryogenesis and in adulthood. Among the cells expressing them are those present in the developing nervous system including neural stem (or progenitor) cells as well as differentiating neurons and oligodendrocytes. SoxD transcription factors do not contain distinct activator or repressor domain, and they are believed to function in modulation of other transcription factors in promoter-specific manners. This brief review article will attempt to summarize the latest studies on the function of SoxD genes in embryogenesis with a particular emphasis on the regulation of neural development.
Collapse
Affiliation(s)
- Eun Hye Ji
- Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Jaesang Kim
- Department of Life Science, Ewha Womans University, Seoul, Korea
| |
Collapse
|
34
|
Buitrago-Delgado E, Nordin K, Rao A, Geary L, LaBonne C. NEURODEVELOPMENT. Shared regulatory programs suggest retention of blastula-stage potential in neural crest cells. Science 2015; 348:1332-5. [PMID: 25931449 PMCID: PMC4652794 DOI: 10.1126/science.aaa3655] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/26/2015] [Indexed: 12/18/2022]
Abstract
Neural crest cells, which are specific to vertebrates, arise in the ectoderm but can generate cell types that are typically categorized as mesodermal. This broad developmental potential persists past the time when most ectoderm-derived cells become lineage-restricted. The ability of neural crest to contribute mesodermal derivatives to the bauplan has raised questions about how this apparent gain in potential is achieved. Here, we describe shared molecular underpinnings of potency in neural crest and blastula cells. We show that in Xenopus, key neural crest regulatory factors are also expressed in blastula animal pole cells and promote pluripotency in both cell types. We suggest that neural crest cells may have evolved as a consequence of a subset of blastula cells retaining activity of the regulatory network underlying pluripotency.
Collapse
Affiliation(s)
- Elsy Buitrago-Delgado
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Kara Nordin
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Anjali Rao
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Lauren Geary
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Carole LaBonne
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA. Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
35
|
Kozhemyakina E, Lassar AB, Zelzer E. A pathway to bone: signaling molecules and transcription factors involved in chondrocyte development and maturation. Development 2015; 142:817-31. [PMID: 25715393 DOI: 10.1242/dev.105536] [Citation(s) in RCA: 387] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Decades of work have identified the signaling pathways that regulate the differentiation of chondrocytes during bone formation, from their initial induction from mesenchymal progenitor cells to their terminal maturation into hypertrophic chondrocytes. Here, we review how multiple signaling molecules, mechanical signals and morphological cell features are integrated to activate a set of key transcription factors that determine and regulate the genetic program that induces chondrogenesis and chondrocyte differentiation. Moreover, we describe recent findings regarding the roles of several signaling pathways in modulating the proliferation and maturation of chondrocytes in the growth plate, which is the 'engine' of bone elongation.
Collapse
Affiliation(s)
- Elena Kozhemyakina
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Building C-Room 305A, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Andrew B Lassar
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Building C-Room 305A, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Elazar Zelzer
- Weizmann Institute of Science, Department of Molecular Genetics, PO Box 26, Rehovot 76100, Israel
| |
Collapse
|