1
|
Dhiman S, Panigrahi I, Sharma M, Chaudhry C, Garg M. TWIST1 Gene Variants Cause Craniosynostosis with Limb Abnormalities in Asian Patients. J Pediatr Genet 2024; 13:258-262. [PMID: 39502847 PMCID: PMC11534420 DOI: 10.1055/s-0043-1771527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 06/25/2023] [Indexed: 11/08/2024]
Abstract
The TWIST1 gene codes for a highly conserved transcription factor in a basic helix-loop-helix transcription factors family. The pattern of inheritance is autosomal dominant in Saethre-Chotzen syndrome, Robinow-Sorauf syndrome, and Sweeney-Cox syndrome. Major features of these syndromes include coronal synostosis, vision problems, and deafness, and facial features include hypertelorism, low-set ears, arched eyebrows, beaked nose, maxillary hypoplasia, and other dysmorphisms including broad great toes, clinodactyly, brachydactyly, and cutaneous syndactyly. TWIST1 (bHLH) transcription factor regulates the formation of head and limbs in the embryo. We describe three families affected with craniosynostosis in whom a sporadic TWIST1 variant was identified on whole exome sequencing, chromosomal microarray, and Sanger sequencing.
Collapse
Affiliation(s)
| | | | - Maryada Sharma
- Department of Otorhinolaryngology, PGIMER, Chandigarh, India
| | | | - Mahak Garg
- Department of Pediatrics, PGIMER, Chandigarh, India
| |
Collapse
|
2
|
Bok S, Sun J, Greenblatt MB. Are osteoblasts multiple cell types? A new diversity in skeletal stem cells and their derivatives. J Bone Miner Res 2024; 39:1386-1392. [PMID: 39052334 PMCID: PMC11425698 DOI: 10.1093/jbmr/zjae109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/08/2024] [Accepted: 05/31/2024] [Indexed: 07/27/2024]
Abstract
Only in the past decade have skeletal stem cells (SSCs), a cell type displaying formal evidence of stemness and serving as the ultimate origin of mature skeletal cell types such as osteoblasts, been defined. Here, we discuss a pair of recent reports that identify that SSCs do not represent a single cell type, but rather a family of related cells that each have characteristic anatomic locations and distinct functions tailored to the physiology of those sites. The distinct functional properties of these SSCs in turn provide a basis for the diseases of their respective locations. This concept emerges from one report identifying a distinct vertebral skeletal stem cell driving the high rate of breast cancer metastasis to the spine over other skeletal sites and a report identifying 2 SSCs in the calvaria that interact to mediate both physiologic calvarial mineralization and pathologic calvarial suture fusion in craniosynostosis. Despite displaying functional differences, these SSCs are each united by shared features including a shared series of surface markers and parallel differentiation hierarchies. We propose that this diversity at the level of SSCs in turn translates into a similar diversity at the level of mature skeletal cell types, including osteoblasts, with osteoblasts derived from different SSCs each displaying different functional and transcriptional characteristics reflecting their cell of origin. In this model, osteoblasts would represent not a single cell type, but rather a family of related cells each with distinct functions, paralleling the functional diversity in SSCs.
Collapse
Affiliation(s)
- Seoyeon Bok
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York NY 10065, United States
| | - Jun Sun
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York NY 10065, United States
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York NY 10065, United States
- Skeletal Health and Orthopedic Research Program, Hospital for Special Surgery, New York NY 10065, United States
| |
Collapse
|
3
|
Tu M, Ge B, Li J, Pan Y, Zhao B, Han J, Wu J, Zhang K, Liu G, Hou M, Yue M, Han X, Sun T, An Y. Emerging biological functions of Twist1 in cell differentiation. Dev Dyn 2024. [PMID: 39254141 DOI: 10.1002/dvdy.736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/03/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024] Open
Abstract
Twist1 is required for embryonic development and expresses after birth in mesenchymal stem cells derived from mesoderm, where it governs mesenchymal cell development. As a well-known regulator of epithelial-mesenchymal transition or embryonic organogenesis, Twist1 is important in a variety of developmental systems, including mesoderm formation, neurogenesis, myogenesis, cranial neural crest cell migration, and differentiation. In this review, we first highlight the physiological significance of Twist1 in cell differentiation, including osteogenic, chondrogenic, and myogenic differentiation, and then detail its probable molecular processes and signaling pathways. On this premise, we summarize the significance of Twist1 in distinct developmental disorders and diseases to provide a reference for studies on cell differentiation/development-related diseases.
Collapse
Affiliation(s)
- Mengjie Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Bingqian Ge
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jiali Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yanbing Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Binbin Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jiayang Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jialin Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Kaifeng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Guangchao Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Mengwen Hou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Man Yue
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Xu Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Tiantian Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| |
Collapse
|
4
|
Wang Y, Qin Q, Wang Z, Negri S, Sono T, Tower RJ, Li Z, Xing X, Archer M, Thottappillil N, Zhu M, Suarez A, Kim DH, Harvey T, Fan CM, James AW. The Mohawk homeobox gene represents a marker and osteo-inhibitory factor in calvarial suture osteoprogenitor cells. Cell Death Dis 2024; 15:420. [PMID: 38886383 PMCID: PMC11183145 DOI: 10.1038/s41419-024-06813-4] [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: 02/03/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024]
Abstract
The regeneration of the mammalian skeleton's craniofacial bones necessitates the action of intrinsic and extrinsic inductive factors from multiple cell types, which function hierarchically and temporally to control the differentiation of osteogenic progenitors. Single-cell transcriptomics of developing mouse calvarial suture recently identified a suture mesenchymal progenitor population with previously unappreciated tendon- or ligament-associated gene expression profile. Here, we developed a Mohawk homeobox (MkxCG; R26RtdT) reporter mouse and demonstrated that this reporter identifies an adult calvarial suture resident cell population that gives rise to calvarial osteoblasts and osteocytes during homeostatic conditions. Single-cell RNA sequencing (scRNA-Seq) data reveal that Mkx+ suture cells display a progenitor-like phenotype with expression of teno-ligamentous genes. Bone injury with Mkx+ cell ablation showed delayed bone healing. Remarkably, Mkx gene played a critical role as an osteo-inhibitory factor in calvarial suture cells, as knockdown or knockout resulted in increased osteogenic differentiation. Localized deletion of Mkx in vivo also resulted in robustly increased calvarial defect repair. We further showed that mechanical stretch dynamically regulates Mkx expression, in turn regulating calvarial cell osteogenesis. Together, we define Mkx+ cells within the suture mesenchyme as a progenitor population for adult craniofacial bone repair, and Mkx acts as a mechanoresponsive gene to prevent osteogenic differentiation within the stem cell niche.
Collapse
Affiliation(s)
- Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Ziyi Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Stefano Negri
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
- Orthopaedic and Trauma Surgery Unit, Department of Surgery, Dentistry, Paediatrics and Gynaecology of the University of Verona, 37134, Verona, Italy
| | - Takashi Sono
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Robert J Tower
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Zhao Li
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Xin Xing
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Mary Archer
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | | | - Manyu Zhu
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Allister Suarez
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Tyler Harvey
- Department of Embryology, Carnegie Institution of Washington, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Chen-Ming Fan
- Department of Embryology, Carnegie Institution of Washington, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
5
|
Arakil N, Akhund SA, Elaasser B, Mohammad KS. Intersecting Paths: Unraveling the Complex Journey of Cancer to Bone Metastasis. Biomedicines 2024; 12:1075. [PMID: 38791037 PMCID: PMC11117796 DOI: 10.3390/biomedicines12051075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 04/27/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
The phenomenon of bone metastases presents a significant challenge within the context of advanced cancer treatments, particularly pertaining to breast, prostate, and lung cancers. These metastatic occurrences stem from the dissemination of cancerous cells into the bone, thereby interrupting the equilibrium between osteoblasts and osteoclasts. Such disruption results in skeletal complications, adversely affecting patient morbidity and quality of life. This review discusses the intricate interplay between cancer cells and the bone microenvironment, positing the bone not merely as a passive recipient of metastatic cells but as an active contributor to cancer progression through its distinctive biochemical and cellular makeup. A thorough examination of bone structure and the dynamics of bone remodeling is undertaken, elucidating how metastatic cancer cells exploit these processes. This review explores the genetic and molecular pathways that underpin the onset and development of bone metastases. Particular emphasis is placed on the roles of cytokines and growth factors in facilitating osteoclastogenesis and influencing osteoblast activity. Additionally, this paper offers a meticulous critique of current diagnostic methodologies, ranging from conventional radiography to advanced molecular imaging techniques, and discusses the implications of a nuanced understanding of bone metastasis biology for therapeutic intervention. This includes the development of targeted therapies and strategies for managing bone pain and other skeletal-related events. Moreover, this review underscores the imperative of ongoing research efforts aimed at identifying novel therapeutic targets and refining management approaches for bone metastases. It advocates for a multidisciplinary strategy that integrates advancements in medical oncology and radiology with insights derived from molecular biology and genetics, to enhance prognostic outcomes and the quality of life for patients afflicted by this debilitating condition. In summary, bone metastases constitute a complex issue that demands a comprehensive and informed approach to treatment. This article contributes to the ongoing discourse by consolidating existing knowledge and identifying avenues for future investigation, with the overarching objective of ameliorating patient care in the domain of oncology.
Collapse
Affiliation(s)
| | | | | | - Khalid S. Mohammad
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 1153, Saudi Arabia; (N.A.); (S.A.A.); (B.E.)
| |
Collapse
|
6
|
Mikheeva SA, Funk CC, Horner PJ, Rostomily RC, Mikheev AM. Novel TCF4:TCF12 heterodimer inhibits glioblastoma growth. Mol Oncol 2024; 18:517-527. [PMID: 37507199 PMCID: PMC10920085 DOI: 10.1002/1878-0261.13496] [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: 02/06/2023] [Revised: 06/15/2023] [Accepted: 07/24/2023] [Indexed: 07/30/2023] Open
Abstract
TWIST1 (TW) is a pro-oncogenic basic helix-loop-helix (bHLH) transcription factor and promotes the hallmark features of malignancy (e.g., cell invasion, cancer cell stemness, and treatment resistance), which contribute to poor prognoses of glioblastoma (GBM). We previously reported that specific TW dimerization motifs regulate unique cellular phenotypes in GBM. For example, the TW:E12 heterodimer increases periostin (POSTN) expression and promotes cell invasion. TW dimer-specific transcriptional regulation requires binding to the regulatory E-box consensus sequences, but alternative bHLH dimers that balance TW dimer activity in regulating pro-oncogenic TW target genes are unknown. We leveraged the ENCODE DNase I hypersensitivity data to identify E-box sites and tethered TW:E12 and TW:TW proteins to validate dimer binding to E-boxes in vitro. Subsequently, TW knockdown revealed a novel TCF4:TCF12 bHLH dimer occupying the same TW E-box site that, when expressed as a tethered TCF4:TCF12 dimer, markedly repressed POSTN expression and extended animal survival. These observations support TCF4:TCF12 as a novel dimer with tumor-suppressor activity in GBM that functions in part through displacement of and/or competitive inhibition of pro-oncogenic TW dimers at E-box sites.
Collapse
Affiliation(s)
- Svetlana A. Mikheeva
- Department of Neurosurgery, Center for NeuroregenerationHouston Methodist Research InstituteTexasUSA
| | - Cory C. Funk
- Institute for Systems BiologySeattleWashingtonUSA
| | - Philip J. Horner
- Department of Neurosurgery, Center for NeuroregenerationHouston Methodist Research InstituteTexasUSA
- Department of NeurosurgeryUniversity of WashingtonSeattleWashingtonUSA
- Institute for Stem Cell and Regenerative MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Robert C. Rostomily
- Department of Neurosurgery, Center for NeuroregenerationHouston Methodist Research InstituteTexasUSA
- Department of NeurosurgeryUniversity of WashingtonSeattleWashingtonUSA
- Institute for Stem Cell and Regenerative MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Andrei M. Mikheev
- Department of Neurosurgery, Center for NeuroregenerationHouston Methodist Research InstituteTexasUSA
- Department of NeurosurgeryUniversity of WashingtonSeattleWashingtonUSA
- Institute for Stem Cell and Regenerative MedicineUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
7
|
Kim S, Morgunova E, Naqvi S, Goovaerts S, Bader M, Koska M, Popov A, Luong C, Pogson A, Swigut T, Claes P, Taipale J, Wysocka J. DNA-guided transcription factor cooperativity shapes face and limb mesenchyme. Cell 2024; 187:692-711.e26. [PMID: 38262408 PMCID: PMC10872279 DOI: 10.1016/j.cell.2023.12.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 10/23/2023] [Accepted: 12/27/2023] [Indexed: 01/25/2024]
Abstract
Transcription factors (TFs) can define distinct cellular identities despite nearly identical DNA-binding specificities. One mechanism for achieving regulatory specificity is DNA-guided TF cooperativity. Although in vitro studies suggest that it may be common, examples of such cooperativity remain scarce in cellular contexts. Here, we demonstrate how "Coordinator," a long DNA motif composed of common motifs bound by many basic helix-loop-helix (bHLH) and homeodomain (HD) TFs, uniquely defines the regulatory regions of embryonic face and limb mesenchyme. Coordinator guides cooperative and selective binding between the bHLH family mesenchymal regulator TWIST1 and a collective of HD factors associated with regional identities in the face and limb. TWIST1 is required for HD binding and open chromatin at Coordinator sites, whereas HD factors stabilize TWIST1 occupancy at Coordinator and titrate it away from HD-independent sites. This cooperativity results in the shared regulation of genes involved in cell-type and positional identities and ultimately shapes facial morphology and evolution.
Collapse
Affiliation(s)
- Seungsoo Kim
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA; Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford, CA 94305, USA
| | - Ekaterina Morgunova
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
| | - Sahin Naqvi
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA; Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Seppe Goovaerts
- Medical Imaging Research Center, UZ Leuven, Leuven, Belgium; Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Maram Bader
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA; Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Mervenaz Koska
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | | | - Christy Luong
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Angela Pogson
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | - Tomek Swigut
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA; Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford, CA 94305, USA
| | - Peter Claes
- Medical Imaging Research Center, UZ Leuven, Leuven, Belgium; Department of Human Genetics, KU Leuven, Leuven, Belgium; Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium
| | - Jussi Taipale
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden; Department of Biochemistry, University of Cambridge, Cambridge, UK; Applied Tumor Genomics Program, University of Helsinki, Helsinki, Finland
| | - Joanna Wysocka
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA; Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford, CA 94305, USA.
| |
Collapse
|
8
|
Bok S, Yallowitz AR, Sun J, McCormick J, Cung M, Hu L, Lalani S, Li Z, Sosa BR, Baumgartner T, Byrne P, Zhang T, Morse KW, Mohamed FF, Ge C, Franceschi RT, Cowling RT, Greenberg BH, Pisapia DJ, Imahiyerobo TA, Lakhani S, Ross ME, Hoffman CE, Debnath S, Greenblatt MB. A multi-stem cell basis for craniosynostosis and calvarial mineralization. Nature 2023; 621:804-812. [PMID: 37730988 PMCID: PMC10799660 DOI: 10.1038/s41586-023-06526-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 08/09/2023] [Indexed: 09/22/2023]
Abstract
Craniosynostosis is a group of disorders of premature calvarial suture fusion. The identity of the calvarial stem cells (CSCs) that produce fusion-driving osteoblasts in craniosynostosis remains poorly understood. Here we show that both physiologic calvarial mineralization and pathologic calvarial fusion in craniosynostosis reflect the interaction of two separate stem cell lineages; a previously identified cathepsin K (CTSK) lineage CSC1 (CTSK+ CSC) and a separate discoidin domain-containing receptor 2 (DDR2) lineage stem cell (DDR2+ CSC) that we identified in this study. Deletion of Twist1, a gene associated with craniosynostosis in humans2,3, solely in CTSK+ CSCs is sufficient to drive craniosynostosis in mice, but the sites that are destined to fuse exhibit an unexpected depletion of CTSK+ CSCs and a corresponding expansion of DDR2+ CSCs, with DDR2+ CSC expansion being a direct maladaptive response to CTSK+ CSC depletion. DDR2+ CSCs display full stemness features, and our results establish the presence of two distinct stem cell lineages in the sutures, with both populations contributing to physiologic calvarial mineralization. DDR2+ CSCs mediate a distinct form of endochondral ossification without the typical haematopoietic marrow formation. Implantation of DDR2+ CSCs into suture sites is sufficient to induce fusion, and this phenotype was prevented by co-transplantation of CTSK+ CSCs. Finally, the human counterparts of DDR2+ CSCs and CTSK+ CSCs display conserved functional properties in xenograft assays. The interaction between these two stem cell populations provides a new biologic interface for the modulation of calvarial mineralization and suture patency.
Collapse
Affiliation(s)
- Seoyeon Bok
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Alisha R Yallowitz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jun Sun
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jason McCormick
- Flow Cytometry Core Facility, Weill Cornell Medicine, New York, NY, USA
| | - Michelle Cung
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lingling Hu
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, NY, USA
| | - Sarfaraz Lalani
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Zan Li
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Branden R Sosa
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Tomas Baumgartner
- Flow Cytometry Core Facility, Weill Cornell Medicine, New York, NY, USA
| | - Paul Byrne
- Flow Cytometry Core Facility, Weill Cornell Medicine, New York, NY, USA
| | - Tuo Zhang
- Genomics Resources Core Facility, Weill Cornell Medicine, New York, NY, USA
| | - Kyle W Morse
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, NY, USA
| | - Fatma F Mohamed
- Department of Periodontics, Prevention and Geriatrics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Chunxi Ge
- Department of Periodontics, Prevention and Geriatrics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Renny T Franceschi
- Department of Periodontics, Prevention and Geriatrics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Randy T Cowling
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, CA, USA
| | - Barry H Greenberg
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, CA, USA
| | - David J Pisapia
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Thomas A Imahiyerobo
- Division of Plastic Surgery, Department of Surgery, New York-Presbyterian Hospital and Columbia University Medical Center, New York, NY, USA
| | - Shenela Lakhani
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - M Elizabeth Ross
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Caitlin E Hoffman
- Department of Neurological Surgery, Weill Cornell Medicine and New York-Presbyterian Hospital, New York, NY, USA
| | - Shawon Debnath
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
- Research Division, Hospital for Special Surgery, New York, NY, USA.
| |
Collapse
|
9
|
Casasnovas-Nieves JJ, Rodríguez Y, Franco HL, Cadilla CL. Mechanisms of Regulation of the CHRDL1 Gene by the TWIST2 and ADD1/SREBP1c Transcription Factors. Genes (Basel) 2023; 14:1733. [PMID: 37761873 PMCID: PMC10530651 DOI: 10.3390/genes14091733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Setleis syndrome (SS) is a rare focal facial dermal dysplasia caused by recessive mutations in the basic helix-loop-helix (bHLH) transcription factor, TWIST2. Expression microarray analysis showed that the chordin-like 1 (CHRDL1) gene is up-regulated in dermal fibroblasts from three SS patients with the Q119X TWIST2 mutation. METHODS Putative TWIST binding sites were found in the upstream region of the CHRDL1 gene and examined by electrophoretic mobility shift (EMSA) and reporter gene assays. RESULTS EMSAs showed specific binding of TWIST1 and TWIST2 homodimers, as well as heterodimers with E12, to the more distal E-boxes. An adjoining E-box was bound by ADD1/SREBP1c. EMSA analysis suggested that TWIST2 and ADD1/SREBP1c could compete for binding. Luciferase (luc) reporter assays revealed that the CHRDL1 gene upstream region drives its expression and ADD1/SREBP1c increased it 2.6 times over basal levels. TWIST2, but not the TWIST2-Q119X mutant, blocked activation by ADD1/SREBP1c, but overexpression of TWIST2-Q119X increased luc gene expression. In addition, EMSA competition assays showed that TWIST2, but not TWIST1, competes with ADD1/SREBP1c for DNA binding to the same site. CONCLUSIONS Formation of an inactive complex between the TWIST2 Q119X and Q65X mutant proteins and ADD1/SREBP1c may prevent repressor binding and allow the binding of other regulators to activate CHRDL1 gene expression.
Collapse
Affiliation(s)
- José J. Casasnovas-Nieves
- Department of Biochemistry, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan 00936, Puerto Rico; (J.J.C.-N.); (Y.R.); (H.L.F.)
| | - Yacidzohara Rodríguez
- Department of Biochemistry, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan 00936, Puerto Rico; (J.J.C.-N.); (Y.R.); (H.L.F.)
| | - Hector L. Franco
- Department of Biochemistry, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan 00936, Puerto Rico; (J.J.C.-N.); (Y.R.); (H.L.F.)
- Department of Genetics, School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, NC 27599, USA
| | - Carmen L. Cadilla
- Department of Biochemistry, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan 00936, Puerto Rico; (J.J.C.-N.); (Y.R.); (H.L.F.)
| |
Collapse
|
10
|
Kim S, Morgunova E, Naqvi S, Bader M, Koska M, Popov A, Luong C, Pogson A, Claes P, Taipale J, Wysocka J. DNA-guided transcription factor cooperativity shapes face and limb mesenchyme. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.29.541540. [PMID: 37398193 PMCID: PMC10312427 DOI: 10.1101/2023.05.29.541540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Transcription factors (TFs) can define distinct cellular identities despite nearly identical DNA-binding specificities. One mechanism for achieving regulatory specificity is DNA-guided TF cooperativity. Although in vitro studies suggest it may be common, examples of such cooperativity remain scarce in cellular contexts. Here, we demonstrate how 'Coordinator', a long DNA motif comprised of common motifs bound by many basic helix-loop-helix (bHLH) and homeodomain (HD) TFs, uniquely defines regulatory regions of embryonic face and limb mesenchyme. Coordinator guides cooperative and selective binding between the bHLH family mesenchymal regulator TWIST1 and a collective of HD factors associated with regional identities in the face and limb. TWIST1 is required for HD binding and open chromatin at Coordinator sites, while HD factors stabilize TWIST1 occupancy at Coordinator and titrate it away from HD-independent sites. This cooperativity results in shared regulation of genes involved in cell-type and positional identities, and ultimately shapes facial morphology and evolution.
Collapse
Affiliation(s)
- Seungsoo Kim
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305
- Department of Developmental Biology, Stanford University, Stanford, CA 94305
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305
- Howard Hughes Medical Institute, Stanford, CA 94305
| | - Ekaterina Morgunova
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
| | - Sahin Naqvi
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305
- Department of Developmental Biology, Stanford University, Stanford, CA 94305
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305
- Department of Genetics, Stanford University, Stanford, CA 94305
| | - Maram Bader
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305
- Department of Developmental Biology, Stanford University, Stanford, CA 94305
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305
| | - Mervenaz Koska
- Department of Developmental Biology, Stanford University, Stanford, CA 94305
| | | | - Christy Luong
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305
| | - Angela Pogson
- Department of Developmental Biology, Stanford University, Stanford, CA 94305
| | - Peter Claes
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium
- Medical Imaging Research Center, UZ Leuven, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Jussi Taipale
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- Applied Tumor Genomics Program, University of Helsinki, Helsinki, Finland
| | - Joanna Wysocka
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305
- Department of Developmental Biology, Stanford University, Stanford, CA 94305
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305
- Howard Hughes Medical Institute, Stanford, CA 94305
| |
Collapse
|
11
|
Hoshino Y, Takechi M, Moazen M, Steacy M, Koyabu D, Furutera T, Ninomiya Y, Nuri T, Pauws E, Iseki S. Synchondrosis fusion contributes to the progression of postnatal craniofacial dysmorphology in syndromic craniosynostosis. J Anat 2023; 242:387-401. [PMID: 36394990 PMCID: PMC9919486 DOI: 10.1111/joa.13790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/16/2022] [Accepted: 10/28/2022] [Indexed: 11/18/2022] Open
Abstract
Syndromic craniosynostosis (CS) patients exhibit early, bony fusion of calvarial sutures and cranial synchondroses, resulting in craniofacial dysmorphology. In this study, we chronologically evaluated skull morphology change after abnormal fusion of the sutures and synchondroses in mouse models of syndromic CS for further understanding of the disease. We found fusion of the inter-sphenoid synchondrosis (ISS) in Apert syndrome model mice (Fgfr2S252W/+ ) around 3 weeks old as seen in Crouzon syndrome model mice (Fgfr2cC342Y/+ ). We then examined ontogenic trajectories of CS mouse models after 3 weeks of age using geometric morphometrics analyses. Antero-ventral growth of the face was affected in Fgfr2S252W/+ and Fgfr2cC342Y/+ mice, while Saethre-Chotzen syndrome model mice (Twist1+/- ) did not show the ISS fusion and exhibited a similar growth pattern to that of control littermates. Further analysis revealed that the coronal suture synostosis in the CS mouse models induces only the brachycephalic phenotype as a shared morphological feature. Although previous studies suggest that the fusion of the facial sutures during neonatal period is associated with midface hypoplasia, the present study suggests that the progressive postnatal fusion of the cranial synchondrosis also contributes to craniofacial dysmorphology in mouse models of syndromic CS. These morphological trajectories increase our understanding of the progression of syndromic CS skull growth.
Collapse
Affiliation(s)
- Yukiko Hoshino
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Office of New Drug V, Pharmaceuticals and Medical Devices Agency (PMDA), Tokyo, Japan
| | - Masaki Takechi
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mehran Moazen
- Department of UCL Mechanical Engineering, University College London, London, UK
| | - Miranda Steacy
- Institute of Child Health, Great Ormond Street, University College London, London, UK
| | - Daisuke Koyabu
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Research and Development Center for Precision Medicine, Tsukuba University, Tsukuba, Japan
| | - Toshiko Furutera
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Youichirou Ninomiya
- Research Organization of Information and Systems, National Institute of Informatics, Tokyo, Japan
| | - Takashi Nuri
- Department of Plastic and Reconstructive Surgery, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Erwin Pauws
- Institute of Child Health, Great Ormond Street, University College London, London, UK
| | - Sachiko Iseki
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
12
|
Auricles Anomalies in Patients With a TCF12 Gene Mutation. J Craniofac Surg 2023; 34:302-305. [PMID: 35994750 DOI: 10.1097/scs.0000000000008938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 06/08/2022] [Indexed: 01/11/2023] Open
Abstract
Craniostenosis is a morphological anomaly affecting about 0.5 of 1000 births and one third of the cases are of genetic origin. Among the syndromes responsible for craniostenosis, there is the Saethre-Chotzen syndrome due to a mutation of the TWIST 1 gene located on chromosome 7. This polymalformative syndrome classically includes a particular morphology of the auricles. The penetrance is variable and results in a phenotypic variability at the origin of "Saethre-Chotzen like" clinical pictures for which the TWIST 1 gene mutation is sometimes not found. Recently, the TCF 12 gene has been implicated in some of these cases. Among the multiple facial malformations, we have carefully examined the particular morphology of the auricle of these patients. The authors found several abnormalities in patients with a TCF 12 gene mutation, namely a thickened and hammered upper pole of the helix, a narrow concha without crux cymbae and a thickened lobe. These morphological features may guide the diagnosis and allow an earlier search for a TCF 12 gene mutation.
Collapse
|
13
|
Ang PS, Matrongolo MJ, Zietowski ML, Nathan SL, Reid RR, Tischfield MA. Cranium growth, patterning and homeostasis. Development 2022; 149:dev201017. [PMID: 36408946 PMCID: PMC9793421 DOI: 10.1242/dev.201017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Craniofacial development requires precise spatiotemporal regulation of multiple signaling pathways that crosstalk to coordinate the growth and patterning of the skull with surrounding tissues. Recent insights into these signaling pathways and previously uncharacterized progenitor cell populations have refined our understanding of skull patterning, bone mineralization and tissue homeostasis. Here, we touch upon classical studies and recent advances with an emphasis on developmental and signaling mechanisms that regulate the osteoblast lineage for the calvaria, which forms the roof of the skull. We highlight studies that illustrate the roles of osteoprogenitor cells and cranial suture-derived stem cells for proper calvarial growth and homeostasis. We also discuss genes and signaling pathways that control suture patency and highlight how perturbing the molecular regulation of these pathways leads to craniosynostosis. Finally, we discuss the recently discovered tissue and signaling interactions that integrate skull and cerebrovascular development, and the potential implications for both cerebrospinal fluid hydrodynamics and brain waste clearance in craniosynostosis.
Collapse
Affiliation(s)
- Phillip S. Ang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
| | - Matt J. Matrongolo
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA
| | | | - Shelby L. Nathan
- Laboratory of Craniofacial Biology and Development, Section of Plastic Surgery, Department of Surgery, University of Chicago Medicine, Chicago, IL 60637, USA
| | - Russell R. Reid
- Laboratory of Craniofacial Biology and Development, Section of Plastic Surgery, Department of Surgery, University of Chicago Medicine, Chicago, IL 60637, USA
| | - Max A. Tischfield
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA
| |
Collapse
|
14
|
Dorafshan S, Razmi M, Safaei S, Gentilin E, Madjd Z, Ghods R. Periostin: biology and function in cancer. Cancer Cell Int 2022; 22:315. [PMID: 36224629 PMCID: PMC9555118 DOI: 10.1186/s12935-022-02714-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 09/13/2022] [Indexed: 11/24/2022] Open
Abstract
Periostin (POSTN), a member of the matricellular protein family, is a secreted adhesion-related protein produced in the periosteum and periodontal ligaments. Matricellular proteins are a nonstructural family of extracellular matrix (ECM) proteins that regulate a wide range of biological processes in both normal and pathological conditions. Recent studies have demonstrated the key roles of these ECM proteins in the tumor microenvironment. Furthermore, periostin is an essential regulator of bone and tooth formation and maintenance, as well as cardiac development. Also, periostin interacts with multiple cell-surface receptors, especially integrins, and triggers signals that promote tumor growth. According to recent studies, these signals are implicated in cancer cell survival, epithelial-mesenchymal transition (EMT), invasion, and metastasis. In this review, we will summarize the most current data regarding periostin, its structure and isoforms, expressions, functions, and regulation in normal and cancerous tissues. Emphasis is placed on its association with cancer progression, and also future potential for periostin-targeted therapeutic approaches will be explored.
Collapse
Affiliation(s)
- Shima Dorafshan
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mahdieh Razmi
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Sadegh Safaei
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Erica Gentilin
- Bioacoustics Research Laboratory, Department of Neurosciences, University of Padua, via G. Orus, 2b, 35129, Padua, Italy
| | - Zahra Madjd
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| | - Roya Ghods
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
15
|
Admoni-Elisha L, Elbaz T, Chopra A, Shapira G, Bedford M, Fry C, Shomron N, Biggar K, Feldman M, Levy D. TWIST1 methylation by SETD6 selectively antagonizes LINC-PINT expression in glioma. Nucleic Acids Res 2022; 50:6903-6918. [PMID: 35694846 PMCID: PMC9262621 DOI: 10.1093/nar/gkac485] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 05/16/2022] [Accepted: 05/30/2022] [Indexed: 12/24/2022] Open
Abstract
Gliomas are one of the most common and lethal brain tumors among adults. One process that contributes to glioma progression and recurrence is the epithelial to mesenchymal transition (EMT). EMT is regulated by a set of defined transcription factors which tightly regulate this process, among them is the basic helix-loop-helix family member, TWIST1. Here we show that TWIST1 is methylated on lysine-33 at chromatin by SETD6, a methyltransferase with expression levels correlating with poor survival in glioma patients. RNA-seq analysis in U251 glioma cells suggested that both SETD6 and TWIST1 regulate cell adhesion and migration processes. We further show that TWIST1 methylation attenuates the expression of the long-non-coding RNA, LINC-PINT, thereby promoting EMT in glioma. Mechanistically, TWIST1 methylation represses the transcription of LINC-PINT by increasing the occupancy of EZH2 and the catalysis of the repressive H3K27me3 mark at the LINC-PINT locus. Under un-methylated conditions, TWIST1 dissociates from the LINC-PINT locus, allowing the expression of LINC-PINT which leads to increased cell adhesion and decreased cell migration. Together, our findings unravel a new mechanistic dimension for selective expression of LINC-PINT mediated by TWIST1 methylation.
Collapse
Affiliation(s)
- Lee Admoni-Elisha
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105 Be'er-Sheva, Israel,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, P.O.B. 653, Be’er-Sheva 84105, Israel
| | - Tzofit Elbaz
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105 Be'er-Sheva, Israel,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, P.O.B. 653, Be’er-Sheva 84105, Israel
| | - Anand Chopra
- Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| | - Guy Shapira
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel,Edmond J. Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv, Israel
| | - Mark T Bedford
- Department of Carcinogenesis, M.D. Anderson Cancer Center, Houston, TX, USA
| | | | - Noam Shomron
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel,Edmond J. Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv, Israel
| | - Kyle Biggar
- Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| | - Michal Feldman
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105 Be'er-Sheva, Israel,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, P.O.B. 653, Be’er-Sheva 84105, Israel
| | - Dan Levy
- To whom correspondence should be addressed. Tel: +972 8 647 7251;
| |
Collapse
|
16
|
Huang Z, Zhang Z, Zhou C, Liu L, Huang C. Epithelial–mesenchymal transition: The history, regulatory mechanism, and cancer therapeutic opportunities. MedComm (Beijing) 2022; 3:e144. [PMID: 35601657 PMCID: PMC9115588 DOI: 10.1002/mco2.144] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 02/05/2023] Open
Abstract
Epithelial–mesenchymal transition (EMT) is a program wherein epithelial cells lose their junctions and polarity while acquiring mesenchymal properties and invasive ability. Originally defined as an embryogenesis event, EMT has been recognized as a crucial process in tumor progression. During EMT, cell–cell junctions and cell–matrix attachments are disrupted, and the cytoskeleton is remodeled to enhance mobility of cells. This transition of phenotype is largely driven by a group of key transcription factors, typically Snail, Twist, and ZEB, through epigenetic repression of epithelial markers, transcriptional activation of matrix metalloproteinases, and reorganization of cytoskeleton. Mechanistically, EMT is orchestrated by multiple pathways, especially those involved in embryogenesis such as TGFβ, Wnt, Hedgehog, and Hippo, suggesting EMT as an intrinsic link between embryonic development and cancer progression. In addition, redox signaling has also emerged as critical EMT modulator. EMT confers cancer cells with increased metastatic potential and drug resistant capacity, which accounts for tumor recurrence in most clinic cases. Thus, targeting EMT can be a therapeutic option providing a chance of cure for cancer patients. Here, we introduce a brief history of EMT and summarize recent advances in understanding EMT mechanisms, as well as highlighting the therapeutic opportunities by targeting EMT in cancer treatment.
Collapse
Affiliation(s)
- Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| | - Chengwei Zhou
- Department of Thoracic Surgery the Affiliated Hospital of Medical School of Ningbo University Ningbo China
| | - Lin Liu
- Department of Thoracic Surgery the Affiliated Hospital of Medical School of Ningbo University Ningbo China
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| |
Collapse
|
17
|
Stanton E, Urata M, Chen JF, Chai Y. The clinical manifestations, molecular mechanisms and treatment of craniosynostosis. Dis Model Mech 2022; 15:dmm049390. [PMID: 35451466 PMCID: PMC9044212 DOI: 10.1242/dmm.049390] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Craniosynostosis is a major congenital craniofacial disorder characterized by the premature fusion of cranial suture(s). Patients with severe craniosynostosis often have impairments in hearing, vision, intracranial pressure and/or neurocognitive functions. Craniosynostosis can result from mutations, chromosomal abnormalities or adverse environmental effects, and can occur in isolation or in association with numerous syndromes. To date, surgical correction remains the primary treatment for craniosynostosis, but it is associated with complications and with the potential for re-synostosis. There is, therefore, a strong unmet need for new therapies. Here, we provide a comprehensive review of our current understanding of craniosynostosis, including typical craniosynostosis types, their clinical manifestations, cranial suture development, and genetic and environmental causes. Based on studies from animal models, we present a framework for understanding the pathogenesis of craniosynostosis, with an emphasis on the loss of postnatal suture mesenchymal stem cells as an emerging disease-driving mechanism. We evaluate emerging treatment options and highlight the potential of mesenchymal stem cell-based suture regeneration as a therapeutic approach for craniosynostosis.
Collapse
Affiliation(s)
- Eloise Stanton
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Mark Urata
- Division of Plastic and Maxillofacial Surgery, Children's Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Jian-Fu Chen
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
18
|
Ting MC, Farmer DT, Teng CS, He J, Chai Y, Crump JG, Maxson RE. Embryonic requirements for Tcf12 in the development of the mouse coronal suture. Development 2022; 149:273884. [PMID: 34878091 PMCID: PMC8783042 DOI: 10.1242/dev.199575] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 11/22/2021] [Indexed: 01/07/2023]
Abstract
A major feature of Saethre-Chotzen syndrome is coronal craniosynostosis, the fusion of the frontal and parietal bones at the coronal suture. It is caused by heterozygous loss-of-function mutations in either of the bHLH transcription factors TWIST1 and TCF12. Although compound heterozygous Tcf12; Twist1 mice display severe coronal synostosis, the individual role of Tcf12 had remained unexplored. Here, we show that Tcf12 controls several key processes in calvarial development, including the rate of frontal and parietal bone growth, and the boundary between sutural and osteogenic cells. Genetic analysis supports an embryonic requirement for Tcf12 in suture formation, as combined deletion of Tcf12 in embryonic neural crest and mesoderm, but not in postnatal suture mesenchyme, disrupts the coronal suture. We also detected asymmetric distribution of mesenchymal cells on opposing sides of the wild-type frontal and parietal bones, which prefigures later bone overlap at the sutures. In Tcf12 mutants, reduced asymmetry is associated with bones meeting end-on-end, possibly contributing to synostosis. Our results support embryonic requirements of Tcf12 in proper formation of the overlapping coronal suture.
Collapse
Affiliation(s)
- Man-chun Ting
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - D'Juan T. Farmer
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Camilla S. Teng
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA,Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jinzhi He
- Center for Craniofacial Molecular Biology, School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| | - J. Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA,Authors for correspondence (, )
| | - Robert E. Maxson
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA,Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA,Authors for correspondence (, )
| |
Collapse
|
19
|
Homodimeric and Heterodimeric Interactions among Vertebrate Basic Helix-Loop-Helix Transcription Factors. Int J Mol Sci 2021; 22:ijms222312855. [PMID: 34884664 PMCID: PMC8657788 DOI: 10.3390/ijms222312855] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/11/2021] [Accepted: 11/17/2021] [Indexed: 01/01/2023] Open
Abstract
The basic helix–loop–helix transcription factor (bHLH TF) family is involved in tissue development, cell differentiation, and disease. These factors have transcriptionally positive, negative, and inactive functions by combining dimeric interactions among family members. The best known bHLH TFs are the E-protein homodimers and heterodimers with the tissue-specific TFs or ID proteins. These cooperative and dynamic interactions result in a complex transcriptional network that helps define the cell’s fate. Here, the reported dimeric interactions of 67 vertebrate bHLH TFs with other family members are summarized in tables, including specifications of the experimental techniques that defined the dimers. The compilation of these extensive data underscores homodimers of tissue-specific bHLH TFs as a central part of the bHLH regulatory network, with relevant positive and negative transcriptional regulatory roles. Furthermore, some sequence-specific TFs can also form transcriptionally inactive heterodimers with each other. The function, classification, and developmental role for all vertebrate bHLH TFs in four major classes are detailed.
Collapse
|
20
|
Romanelli Tavares VL, Guimarães-Ramos SL, Zhou Y, Masotti C, Ezquina S, Moreira DDP, Buermans H, Freitas RS, Den Dunnen JT, Twigg SRF, Passos-Bueno MR. New locus underlying auriculocondylar syndrome (ARCND): 430 kb duplication involving TWIST1 regulatory elements. J Med Genet 2021; 59:895-905. [PMID: 34750192 PMCID: PMC9411924 DOI: 10.1136/jmedgenet-2021-107825] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 09/29/2021] [Indexed: 11/13/2022]
Abstract
Background Auriculocondylar syndrome (ARCND) is a rare genetic disease that affects structures derived from the first and second pharyngeal arches, mainly resulting in micrognathia and auricular malformations. To date, pathogenic variants have been identified in three genes involved in the EDN1-DLX5/6 pathway (PLCB4, GNAI3 and EDN1) and some cases remain unsolved. Here we studied a large unsolved four-generation family. Methods We performed linkage analysis, resequencing and Capture-C to investigate the causative variant of this family. To test the pathogenicity of the CNV found, we modelled the disease in patient craniofacial progenitor cells, including induced pluripotent cell (iPSC)-derived neural crest and mesenchymal cells. Results This study highlights a fourth locus causative of ARCND, represented by a tandem duplication of 430 kb in a candidate region on chromosome 7 defined by linkage analysis. This duplication segregates with the disease in the family (LOD score=2.88) and includes HDAC9, which is located over 200 kb telomeric to the top candidate gene TWIST1. Notably, Capture-C analysis revealed multiple cis interactions between the TWIST1 promoter and possible regulatory elements within the duplicated region. Modelling of the disease revealed an increased expression of HDAC9 and its neighbouring gene, TWIST1, in neural crest cells. We also identified decreased migration of iPSC-derived neural crest cells together with dysregulation of osteogenic differentiation in iPSC-affected mesenchymal stem cells. Conclusion Our findings support the hypothesis that the 430 kb duplication is causative of the ARCND phenotype in this family and that deregulation of TWIST1 expression during craniofacial development can contribute to the phenotype.
Collapse
Affiliation(s)
| | | | - Yan Zhou
- Clinical Genetics Group, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Cibele Masotti
- Genética e Biologia Evolutiva, Universidade de São Paulo Instituto de Biociências, Sao Paulo, Brazil.,Molecular Oncology Center, Hospital Sírio-Libanês, Sao Paulo, Brazil
| | - Suzana Ezquina
- Genética e Biologia Evolutiva, Universidade de São Paulo Instituto de Biociências, Sao Paulo, Brazil.,Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Danielle de Paula Moreira
- Genética e Biologia Evolutiva, Universidade de São Paulo Instituto de Biociências, Sao Paulo, Brazil
| | - Henk Buermans
- Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Renato S Freitas
- Centro de Atendimento Integral ao Fissurado Lábio Palatal, Curitiba, Brazil
| | - Johan T Den Dunnen
- Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Stephen R F Twigg
- Clinical Genetics Group, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Maria Rita Passos-Bueno
- Genética e Biologia Evolutiva, Universidade de São Paulo Instituto de Biociências, Sao Paulo, Brazil
| |
Collapse
|
21
|
Jacob T, Chakravarty A, Panchal A, Patil M, Ghodadra G, Sudhakaran J, Nuesslein-Volhard C. Zebrafish twist2/dermo1 regulates scale shape and scale organization during skin development and regeneration. Cells Dev 2021; 166:203684. [PMID: 33994357 DOI: 10.1016/j.cdev.2021.203684] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 12/22/2020] [Accepted: 04/26/2021] [Indexed: 01/01/2023]
Abstract
Scales are skin appendages in fishes that evolutionarily predate feathers in birds and hair in mammals. Zebrafish scales are dermal in origin and develop during metamorphosis. Understanding regulation of scale development in zebrafish offers an exciting possibility of unraveling how the mechanisms of skin appendage formation evolved in lower vertebrates and whether these mechanisms remained conserved in birds and mammals. Here we have investigated the expression and function of twist 2/dermo1 gene - known for its function in feather and hair formation - in scale development and regeneration. We show that of the four zebrafish twist paralogues, twist2/dermo1 and twist3 are expressed in the scale forming cells during scale development. Their expression is also upregulated during scale regeneration. Our knockout analysis reveals that twist2/dermo1 gene functions in the maintenance of the scale shape and organization during development as well as regeneration. We further show that the expression of twist2/dermo1 and twist3 is regulated by Wnt signaling. Our results demonstrate that the function of twist2/dermo1 in skin appendage formation, presumably under regulation of Wnt signaling, originated during evolution of basal vertebrates.
Collapse
Affiliation(s)
- Tressa Jacob
- Department of Biology, Indian Institute of Science Education and Research-Pune (IISER-Pune), Dr Homi Bhabha Road, Pashan, Pune 411008, India.
| | - Antara Chakravarty
- Department of Biology, Indian Institute of Science Education and Research-Pune (IISER-Pune), Dr Homi Bhabha Road, Pashan, Pune 411008, India
| | - Ankita Panchal
- Dr. Vikram Sarabhai Institute of Cell and Molecular Biology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Gujarat, India
| | - Manjiri Patil
- Department of Biology, Indian Institute of Science Education and Research-Pune (IISER-Pune), Dr Homi Bhabha Road, Pashan, Pune 411008, India
| | - Gautami Ghodadra
- Dr. Vikram Sarabhai Institute of Cell and Molecular Biology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Gujarat, India
| | - Jyotish Sudhakaran
- Department of Biology, Indian Institute of Science Education and Research-Pune (IISER-Pune), Dr Homi Bhabha Road, Pashan, Pune 411008, India
| | | |
Collapse
|
22
|
Chen F, Hua J, Shen H, Wang H. Effect of TWIST1 Gene on the Proliferation and Apoptosis of Human Glioma Cell Line TJ861 by Regulating Mammalian Target of Rapamycin Signaling Pathway. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
To observe TWIST1 gene expression in human glioma and study the effect human glioma cell line TJ861 on the proliferation and apoptosis, and further explore its potential mechanism to provide some reference for the targeted treatment of glioma in the future. Detection of cancer tissue
(Carcinoma tissue) in 55 patients with glioma by RT-PCR and Expression level of TWIST1 in normal and paracancerous tissues (Adjacent tissue), the human glioma cell line TJ861 was further divided into, Nonsense sequence group, (si-NS group), TWIST1 Inhibition group (si-TWIST1 group) and control
group. The glioma cells of si-NS group and si-TWIST1 group were transfected with nonsense sequence and TWIST1 siRNA respectively by liposome transfection technology. Use CCK8 assay to test the cell proliferation ability of each group at 0, 12, 24, 36, 48 and 72 hours; 48 hours after siRNA
transfection, The ability of DNA replication in each group was detected by EdU staining; Apoptosis related protein expression, in each group, was analyzed by Western blot; TUNEL staining was used to test the apoptosis rate of each group; In the end, We studied TWIST1 effect knocking down on
mTOR protein expression in human glioma cells and mTOR protein expression in cancer and adjacent tissues. TWIST1 expression in glioma cells was higher, compared with normal tissues (P <0.05); After transfection of TWIST1 siRNA into human glioma cell line TJ861 in vitro, CCK8
showed glioma cells proliferation ability in si-TWIST1 group at 12, 24, 36, 48 and 72 hours was lower, compared with the control group (P <0.05); After siRNA transfection at 48 hours, the DNA replication ability of glioma cells decreased significantly (P <0.05) with EdU
staining; The inhibition of TWIST1 increased Bax expression in glioma cells, and inhibited Bcl-2 expression (P < 0.05) with Western blot; TUNEL staining further confirmed that the apoptosis level of glioma cells in the si-TWIST1 group was higher, compared with the control group (P
<0.05). Finally, we found that mTOR protein expression in glioma was higher, compared with adjacent tissues. in vitro experiments showed that mTOR expression in glioma cells was decreased after the inhibition of TWIST1 (P <0.05). TWIST1 expression level in glioma was increased.
The inhibition of TWIST1 inhibits the proliferation of glioma by blocking the mTOR signal pathway, and promote the apoptosis of glioma.
Collapse
Affiliation(s)
- Fei Chen
- Department of Neurosurgery, The Sixth People’s Hospital of Nantong, Nantong, Jiangsu, 226011, China
| | - Jiajia Hua
- Traditional Chinese Medicine Department, The Sixth People’s Hospital of Nantong, Nantong, Jiangsu, 226011, China
| | - HongWei Shen
- Health Management Centre, The Sixth People’s Hospital of Shanghai (The East Part), Shanghai, 201306, China
| | - HongLiang Wang
- Department of Neurosurgery, The Sixth People’s Hospital of Nantong, Nantong, Jiangsu, 226011, China
| |
Collapse
|
23
|
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: 17] [Impact Index Per Article: 5.7] [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
|
24
|
Pribadi C, Camp E, Cakouros D, Anderson P, Glackin C, Gronthos S. Pharmacological targeting of KDM6A and KDM6B, as a novel therapeutic strategy for treating craniosynostosis in Saethre-Chotzen syndrome. Stem Cell Res Ther 2020; 11:529. [PMID: 33298158 PMCID: PMC7726873 DOI: 10.1186/s13287-020-02051-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 11/26/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND During development, excessive osteogenic differentiation of mesenchymal progenitor cells (MPC) within the cranial sutures can lead to premature suture fusion or craniosynostosis, leading to craniofacial and cognitive issues. Saethre-Chotzen syndrome (SCS) is a common form of craniosynostosis, caused by TWIST-1 gene mutations. Currently, the only treatment option for craniosynostosis involves multiple invasive cranial surgeries, which can lead to serious complications. METHODS The present study utilized Twist-1 haploinsufficient (Twist-1del/+) mice as SCS mouse model to investigate the inhibition of Kdm6a and Kdm6b activity using the pharmacological inhibitor, GSK-J4, on calvarial cell osteogenic potential. RESULTS This study showed that the histone methyltransferase EZH2, an osteogenesis inhibitor, is downregulated in calvarial cells derived from Twist-1del/+ mice, whereas the counter histone demethylases, Kdm6a and Kdm6b, known promoters of osteogenesis, were upregulated. In vitro studies confirmed that siRNA-mediated inhibition of Kdm6a and Kdm6b expression suppressed osteogenic differentiation of Twist-1del/+ calvarial cells. Moreover, pharmacological targeting of Kdm6a and Kdm6b activity, with the inhibitor, GSK-J4, caused a dose-dependent suppression of osteogenic differentiation by Twist-1del/+ calvarial cells in vitro and reduced mineralized bone formation in Twist-1del/+ calvarial explant cultures. Chromatin immunoprecipitation and Western blot analyses found that GSK-J4 treatment elevated the levels of the Kdm6a and Kdm6b epigenetic target, the repressive mark of tri-methylated lysine 27 on histone 3, on osteogenic genes leading to repression of Runx2 and Alkaline Phosphatase expression. Pre-clinical in vivo studies showed that local administration of GSK-J4 to the calvaria of Twist-1del/+ mice prevented premature suture fusion and kept the sutures open up to postnatal day 20. CONCLUSION The inhibition of Kdm6a and Kdm6b activity by GSK-J4 could be used as a potential non-invasive therapeutic strategy for preventing craniosynostosis in children with SCS. Pharmacological targeting of Kdm6a/b activity can alleviate craniosynostosis in Saethre-Chotzen syndrome. Aberrant osteogenesis by Twist-1 mutant cranial suture mesenchymal progenitor cells occurs via deregulation of epigenetic modifiers Ezh2 and Kdm6a/Kdm6b. Suppression of Kdm6a- and Kdm6b-mediated osteogenesis with GSK-J4 inhibitor can prevent prefusion of cranial sutures.
Collapse
Affiliation(s)
- Clara Pribadi
- Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia.,Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Esther Camp
- Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia.,Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Dimitrios Cakouros
- Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia.,Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Peter Anderson
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Craniofacial Unit, Women and Children Hospital, North Adelaide, South Australia, Australia
| | - Carlotta Glackin
- Molecular Medicine and Neurosciences, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia. .,Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.
| |
Collapse
|
25
|
Ertosun MG, PehlİvanoĞlu S, DİlmaÇ S, TanriÖver G, ÖzeŞ ON. AKT-mediated phosphorylation of TWIST1 is essential for breast cancer cell metastasis. ACTA ACUST UNITED AC 2020; 44:158-165. [PMID: 32922123 PMCID: PMC7478131 DOI: 10.3906/biy-1912-74] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Previously, it was shown that human TWIST1 (basic helix-loop-helix (b-HLH) is phosphorylated by Akt kinase at S42, T121, and S123. To show in vivo effect of these phosphorylations, we created mouse TWIST1 expression vector and converted the codons of S42, T125, and S127 to unphosphorylatable alanine and phosphorylation mimicking Glutamic acid. We hypothesized that alanine mutants would inhibit the metastatic ability of 4T1 cells while glutamic acid mutants would convert nonmetastatic 67NR cells into metastatic phenotype. To confirm this hypothesis, we created metastatic 4T1 and nonmetastatic 67NR cells expressing alanine mutants and glutamic acid mutants mouse TWIST1, respectively. Then, we injected 1 × 106 67NR and 1 × 105 4T1 cells overexpressing mutants of TWIST1 into the breast tissue of BALB/c mice. At the end of the 4th week, we sacrificed the animals, determined the numbers of tumors at lungs and liver. Although 67NR cells overexpressing wild-type TWIST1 did not show any metastasis, cells overexpressing S42E and T125E mutants showed 15–30 macroscopic metastasis to liver and lungs. Parallel to this, 4T1 cells expressing S42A and T125A mutants of TWIST1 showed no macroscopic metastasis. Our results indicate that phosphorylation of S42 and T125 by AKT is essential for TWIST1-mediated tumor growth and metastasis.
Collapse
Affiliation(s)
- Mustafa Gökhan Ertosun
- Department Plastic, Reconstructive and Aesthetic Surgery, Faculty of Medicine, Akdeniz University, Antalya Turkey
| | - Suray PehlİvanoĞlu
- Department of Molecular Biology, Faculty of Science, Necmettin Erbakan University, Konya Turkey
| | - Sayra DİlmaÇ
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya Turkey
| | - Gamze TanriÖver
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya Turkey
| | | |
Collapse
|
26
|
Holmes G, Gonzalez-Reiche AS, Lu N, Zhou X, Rivera J, Kriti D, Sebra R, Williams AA, Donovan MJ, Potter SS, Pinto D, Zhang B, van Bakel H, Jabs EW. Integrated Transcriptome and Network Analysis Reveals Spatiotemporal Dynamics of Calvarial Suturogenesis. Cell Rep 2020; 32:107871. [PMID: 32640236 PMCID: PMC7379176 DOI: 10.1016/j.celrep.2020.107871] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/14/2020] [Accepted: 06/15/2020] [Indexed: 11/28/2022] Open
Abstract
Craniofacial abnormalities often involve sutures, the growth centers of the skull. To characterize the organization and processes governing their development, we profile the murine frontal suture, a model for sutural growth and fusion, at the tissue- and single-cell level on embryonic days (E)16.5 and E18.5. For the wild-type suture, bulk RNA sequencing (RNA-seq) analysis identifies mesenchyme-, osteogenic front-, and stage-enriched genes and biological processes, as well as alternative splicing events modifying the extracellular matrix. Single-cell RNA-seq analysis distinguishes multiple subpopulations, of which five define a mesenchyme-osteoblast differentiation trajectory and show variation along the anteroposterior axis. Similar analyses of in vivo mouse models of impaired frontal suturogenesis in Saethre-Chotzen and Apert syndromes, Twist1+/- and Fgfr2+/S252W, demonstrate distinct transcriptional changes involving angiogenesis and ribogenesis, respectively. Co-expression network analysis reveals gene expression modules from which we validate key driver genes regulating osteoblast differentiation. Our study provides a global approach to gain insights into suturogenesis.
Collapse
Affiliation(s)
- Greg Holmes
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Ana S Gonzalez-Reiche
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Na Lu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xianxiao Zhou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joshua Rivera
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Divya Kriti
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Anthony A Williams
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michael J Donovan
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - S Steven Potter
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, OH 45229, USA
| | - Dalila Pinto
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, and Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Harm van Bakel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Ethylin Wang Jabs
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Cell, Developmental and Regenerative Biology and Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
27
|
TWIST1 Homodimers and Heterodimers Orchestrate Lineage-Specific Differentiation. Mol Cell Biol 2020; 40:MCB.00663-19. [PMID: 32179550 DOI: 10.1128/mcb.00663-19] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 02/27/2020] [Indexed: 01/09/2023] Open
Abstract
The extensive array of basic helix-loop-helix (bHLH) transcription factors and their combinations as dimers underpin the diversity of molecular function required for cell type specification during embryogenesis. The bHLH factor TWIST1 plays pleiotropic roles during development. However, which combinations of TWIST1 dimers are involved and what impact each dimer imposes on the gene regulation network controlled by TWIST1 remain elusive. In this work, proteomic profiling of human TWIST1-expressing cell lines and transcriptome analysis of mouse cranial mesenchyme have revealed that TWIST1 homodimers and heterodimers with TCF3, TCF4, and TCF12 E-proteins are the predominant dimer combinations. Disease-causing mutations in TWIST1 can impact dimer formation or shift the balance of different types of TWIST1 dimers in the cell, which may underpin the defective differentiation of the craniofacial mesenchyme. Functional analyses of the loss and gain of TWIST1-E-protein dimer activity have revealed previously unappreciated roles in guiding lineage differentiation of embryonic stem cells: TWIST1-E-protein heterodimers activate the differentiation of mesoderm and neural crest cells, which is accompanied by the epithelial-to-mesenchymal transition. At the same time, TWIST1 homodimers maintain the stem cells in a progenitor state and block entry to the endoderm lineage.
Collapse
|
28
|
Vincentz JW, Clouthier DE, Firulli AB. Mis-Expression of a Cranial Neural Crest Cell-Specific Gene Program in Cardiac Neural Crest Cells Modulates HAND Factor Expression, Causing Cardiac Outflow Tract Phenotypes. J Cardiovasc Dev Dis 2020; 7:jcdd7020013. [PMID: 32325975 PMCID: PMC7344951 DOI: 10.3390/jcdd7020013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 04/14/2020] [Indexed: 12/22/2022] Open
Abstract
Congenital heart defects (CHDs) occur with such a frequency that they constitute a significant cause of morbidity and mortality in both children and adults. A significant portion of CHDs can be attributed to aberrant development of the cardiac outflow tract (OFT), and of one of its cellular progenitors known as the cardiac neural crest cells (NCCs). The gene regulatory networks that identify cardiac NCCs as a distinct NCC population are not completely understood. Heart and neural crest derivatives (HAND) bHLH transcription factors play essential roles in NCC morphogenesis. The Hand1PA/OFT enhancer is dependent upon bone morphogenic protein (BMP) signaling in both cranial and cardiac NCCs. The Hand1PA/OFT enhancer is directly repressed by the endothelin-induced transcription factors DLX5 and DLX6 in cranial but not cardiac NCCs. This transcriptional distinction offers the unique opportunity to interrogate NCC specification, and to understand why, despite similarities, cranial NCC fate determination is so diverse. We generated a conditionally active transgene that can ectopically express DLX5 within the developing mouse embryo in a Cre-recombinase-dependent manner. Ectopic DLX5 expression represses cranial NCC Hand1PA/OFT-lacZ reporter expression more effectively than cardiac NCC reporter expression. Ectopic DLX5 expression induces broad domains of NCC cell death within the cranial pharyngeal arches, but minimal cell death in cardiac NCC populations. This study shows that transcription control of NCC gene regulatory programs is influenced by their initial specification at the dorsal neural tube.
Collapse
Affiliation(s)
- Joshua W. Vincentz
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics, Anatomy and Medical and Molecular Genetics, Indiana Medical School, Indianapolis, IN 46202, USA
- Correspondence: (J.W.V.); (A.B.F.)
| | - David E. Clouthier
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Anthony B. Firulli
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics, Anatomy and Medical and Molecular Genetics, Indiana Medical School, Indianapolis, IN 46202, USA
- Correspondence: (J.W.V.); (A.B.F.)
| |
Collapse
|
29
|
An ENU-induced mutation in Twist1 transactivation domain causes hindlimb polydactyly with complete penetrance and dominant-negatively impairs E2A-dependent transcription. Sci Rep 2020; 10:2501. [PMID: 32051525 PMCID: PMC7016005 DOI: 10.1038/s41598-020-59455-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 01/29/2020] [Indexed: 11/08/2022] Open
Abstract
Twist1 encodes a basic helix-loop-helix transcription factor (TF), which forms homodimer or heterodimer with other TFs, like E2A, to regulate target genes' expression. Mutations in TWIST1 are associated with Saethre-Chotzen syndrome (SCS), a rare congenital disorder characterized with osteogenesis abnormalities. However, how dysfunction of TWIST1 leads to SCS is still largely unknown. Here, using an unbiased ENU-induced mutagenesis screening, we identified a novel Twist1 mutation and the mutant mouse phenocopies some features of SCS in a dominant manner. Physically, our mutation p.F191S lies at the edge of a predicted α-helix in Twist1 transactivation (TA) domain. Adjacent to F191, a consecutive three-residue (AFS) has been hit by 3 human and 2 mouse disease-associated mutations, including ours. Unlike previously reported mouse null and p.S192P alleles that lead to hindlimb polydactyly with incomplete penetrance but a severe craniofacial malformation, our p.F191S causes the polydactyly (84.2% bilateral and 15.8% unilateral) with complete penetrance but a mild craniofacial malformation. Consistent with the higher penetrance, p.F191S has stronger impairment on E2A-dependent transcription than p.S192P. Although human p.A186T and mouse p.S192P disease mutations are adjacent to ours, these three mutations function differently to impair the E2A-dependent transcription. Unlike p.A186T and p.S192S that disturb local protein conformation and unstabilize the mutant proteins, p.F191S keeps the mutant protein stable and its interaction with E2A entire. Therefore, we argue that p.F191S we identified acts in a dominant-negative manner to impair E2A-dependent transcription and to cause the biological consequences. In addition, the mutant mouse we provided here could be an additional and valuable model for better understanding the disease mechanisms underlying SCS caused by TWIST1 dysfunction.
Collapse
|
30
|
Partially Penetrant Cardiac Neural Crest Defects in Hand1 Phosphomutant Mice: Dimer Choice That Is Not So Critical. Pediatr Cardiol 2019; 40:1339-1344. [PMID: 31338559 PMCID: PMC6786956 DOI: 10.1007/s00246-019-02162-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 07/16/2019] [Indexed: 10/26/2022]
Abstract
Hand1 is a basic Helix-loop-Helix transcription factor that exhibits post-translationally regulated dimer partner choice that allows for a diverse set of Hand1 transcriptional complexes. Indeed, when Hand1 phosphoregulation is altered, conditionally activated hypophorylation (Hand1PO4-) and phosphorylation mimic (Hand1PO4+) Hand1 alleles disrupt both craniofacial and limb morphogenesis with 100% penetrance. Interestingly, activation of conditional Hand1 Phosphomutant alleles within post-migratory neural crest cells produce heart defects that include ventricular septal defects, double-outlet right ventricle, persistent truncus arteriosus with partial penetrance. Single versus double-lobed thymus is a distinguishing feature between Wnt1-Cre;Hand1PO4-/+ and Wnt1-Cre;Hand1PO4+/+ mice. These data show that although Hand1 dimer regulation plays critical and consistent roles in disrupting craniofacial and limb morphogenesis, Hand1 dimer regulation during cardiac outflow track formation is less critical for normal morphogenesis. This review will present the OFT phenotypes observed in Hand1 Phosphomutant mice, and discuss possible mechanisms of how penetrance differences within the same tissues within the same embryos could be variable.
Collapse
|
31
|
TWIST1 Heterodimerization with E12 Requires Coordinated Protein Phosphorylation to Regulate Periostin Expression. Cancers (Basel) 2019; 11:cancers11091392. [PMID: 31540485 PMCID: PMC6770789 DOI: 10.3390/cancers11091392] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 09/04/2019] [Accepted: 09/15/2019] [Indexed: 11/16/2022] Open
Abstract
Diffuse invasion into adjacent brain matter by glioblastoma (GBM) is largely responsible for their dismal prognosis. Previously, we showed that the TWIST1 (TW) bHLH transcription factor and its regulated gene periostin (POSTN) promote invasive phenotypes of GBM cells. Since TW functional effects are regulated by phosphorylation and dimerization, we investigated how phosphorylation of serine 68 in TW regulates TW dimerization, POSTN expression, and invasion in glioma cells. Compared with wild-type TW, the hypophosphorylation mutant, TW(S68A), impaired TW heterodimerization with the E12 bHLH transcription factor and cell invasion in vitro but had no effect on TW homodimerization. Overexpression of TW:E12 forced dimerization constructs (FDCs) increased glioma cell invasion and upregulated pro-invasive proteins, including POSTN, in concert with cytoskeletal reorganization. By contrast, TW:TW homodimer FDCs inhibited POSTN expression and cell invasion in vitro. Further, phosphorylation of analogous PXSP phosphorylation sites in TW:E12 FDCs (TW S68 and E12 S139) coordinately regulated POSTN and PDGFRa mRNA expression. These results suggested that TW regulates pro-invasive phenotypes in part through coordinated phosphorylation events in TW and E12 that promote heterodimer formation and regulate downstream targets. This new mechanistic understanding provides potential therapeutic strategies to inhibit TW-POSTN signaling in GBM and other cancers.
Collapse
|
32
|
Yilmaz E, Mihci E, Nur B, Alper OM. Coronal craniosynostosis due to TCF12 mutations in patients from Turkey. Am J Med Genet A 2019; 179:2241-2245. [PMID: 31353793 DOI: 10.1002/ajmg.a.61311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 06/13/2019] [Accepted: 07/18/2019] [Indexed: 12/30/2022]
Abstract
Craniosynostosis consists of premature fusion of one or more cranial sutures and can be seen as part of a syndrome or diagnosed as nonsyndromic (isolated). Although more than 180 craniosynostosis syndromes have been identified, 70% of the cases are diagnosed as nonsyndromic. On the other hand, genetic causes of the cases are mostly unknown and the overall frequency of the genetic diagnosis is around 25%. In this study, we used targeted Next Generation Sequencing (NGS) analysis to identify the genetic variations of two craniosynostosis cases. We have identified two different truncating mutations, a known NM_207036.1:c.778_779delAT;p.(Met260Valfs*5) and a novel NM_207036.1:c.1102_1108delTCACCTC;p.(Pro369Glnfs*26) TCF12 variants. Additionally, upon physical examination of these two cases, we have observed some shared clinical similarities as well as differences such as bilateral simian crease and hidden cleft palate. This is the first study that reports the TCF12 mutations in Turkish patients with coronal suture synostosis.
Collapse
Affiliation(s)
- Elanur Yilmaz
- Department of Medical Biology and Genetics, Akdeniz University Medical School, Antalya, Turkey
| | - Ercan Mihci
- Department of Pediatric Genetics, Akdeniz University Medical School, Antalya, Turkey
| | - Banu Nur
- Department of Pediatric Genetics, Akdeniz University Medical School, Antalya, Turkey
| | - Ozgul M Alper
- Department of Medical Biology and Genetics, Akdeniz University Medical School, Antalya, Turkey
| |
Collapse
|
33
|
Downregulation of miR-224-5p Promotes Migration and Proliferation in Human Dental Pulp Stem Cells. BIOMED RESEARCH INTERNATIONAL 2019; 2019:4759060. [PMID: 31396530 PMCID: PMC6668552 DOI: 10.1155/2019/4759060] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 06/25/2019] [Indexed: 12/22/2022]
Abstract
Introduction Pulp regeneration, as a treatment for pulp necrosis, has significant advantages over root canal therapy for the preservation of living pulp. To date, research on pulp regeneration has mainly focused on the transplantation of pulp stem cells into the root canal, but there is still a lack of research on the migration of pulp cells into the root canal via cell homing. Stem cells from the apical tooth papilla (SCAP) are recognized as multidirectional stem cells, but these cells are difficult to obtain. MicroRNAs are small noncoding RNAs that play crucial roles in regulating normal and pathologic functions. We hypothesized that some types of microRNAs might improve the migration and proliferation function of dental pulp stem cells (DPSCs), which are easily obtained in clinical practice, and as a result, DPSCs might replace SCAP and provide valuable information for regenerative endodontics. Methods Magnetic activated cell sorting of DPSCs and SCAP was performed. Next-generation sequencing was performed to examine DPSCs and SCAP miRNAs expression and to identify the most significant differentially expressed miRNA. CCK-8 and transwell assays were used to determine the impact of this miRNA on DPSCs proliferation and migration. Results The most significant differentially expressed miRNA between DPSCs and SCAP was miR-224-5p. Downregulating miR-224-5p promoted DPSCs proliferation and migration; the opposite results were observed when miR-224-5p was upregulated. Conclusion MiR-224-5p promotes proliferation and migration in DPSCs, a finding that is of great significance for further exploring the role of dental pulp stem cells in regenerative endodontics.
Collapse
|
34
|
On the traces of tcf12: Investigation of the gene expression pattern during development and cranial suture patterning in zebrafish (Danio rerio). PLoS One 2019; 14:e0218286. [PMID: 31188878 PMCID: PMC6561585 DOI: 10.1371/journal.pone.0218286] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/29/2019] [Indexed: 12/11/2022] Open
Abstract
The transcription factor 12 (tcf12) is a basic Helix-Loop-Helix protein (bHLH) of the E-protein family, proven to play an important role in developmental processes like neurogenesis, mesoderm formation, and cranial vault development. In humans, mutations in TCF12 lead to craniosynostosis, a congenital birth disorder characterized by the premature fusion of one or several of the cranial sutures. Current research has been primarily focused on functional studies of TCF12, hence the cellular expression profile of this gene during embryonic development and early stages of ossification remains poorly understood. Here we present the establishment and detailed analysis of two transgenic tcf12:EGFP fluorescent zebrafish (Danio rerio) reporter lines. Using these transgenic lines, we analyzed the general spatiotemporal expression pattern of tcf12 during different developmental stages and put emphasis on skeletal development and cranial suture patterning. We identified robust tcf12 promoter-driven EGFP expression in the central nervous system (CNS), the heart, the pronephros, and the somites of zebrafish embryos. Additionally, expression was observed inside the muscles and bones of the viscerocranium in juvenile and adult fish. During cranial vault development, the transgenic fish show a high amount of tcf12 expressing cells at the growth fronts of the ossifying frontal and parietal bones and inside the emerging cranial sutures. Subsequently, we tested the transcriptional activity of three evolutionary conserved non-coding elements (CNEs) located in the tcf12 locus by transient transgenic assays and compared their in vivo activity to the expression pattern determined in the transgenic tcf12:EGFP lines. We could validate two of them as tcf12 enhancer elements driving specific gene expression in the CNS during embryogenesis. Our newly established transgenic lines enhance the understanding of tcf12 gene regulation and open up the possibilities for further functional investigation of these novel tcf12 enhancer elements in zebrafish.
Collapse
|
35
|
The Structure of the Periostin Gene, Its Transcriptional Control and Alternative Splicing, and Protein Expression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1132:7-20. [PMID: 31037620 DOI: 10.1007/978-981-13-6657-4_2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Although many studies have described the role of periostin in various diseases, the functions of periostin derived from alternative splicing and proteinase cleavage at its C-terminus remain unknown. Further experiments investigating the periostin structures that are relevant to diseases are essential for an in-depth understanding of their functions, which would accelerate their clinical applications by establishing new approaches for curing intractable diseases. Furthermore, this understanding would enhance our knowledge of novel functions of periostin related to stemness and response to mechanical stress .
Collapse
|
36
|
Wu X, Gu Y. Signaling Mechanisms Underlying Genetic Pathophysiology of Craniosynostosis. Int J Biol Sci 2019; 15:298-311. [PMID: 30745822 PMCID: PMC6367540 DOI: 10.7150/ijbs.29183] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 11/30/2018] [Indexed: 12/14/2022] Open
Abstract
Craniosynostosis, is the premature fusion of one or more cranial sutures which is the second most common cranial facial anomalies. The premature cranial sutures leads to deformity of skull shape and restricts the growth of brain, which might elicit severe neurologic damage. Craniosynostosis exhibit close correlations with a varieties of syndromes. During the past two decades, as the appliance of high throughput DNA sequencing techniques, steady progresses has been made in identifying gene mutations in both syndromic and nonsyndromic cases, which allow researchers to better understanding the genetic roles in the development of cranial vault. As the enrichment of known mutations involved in the pathogenic of premature sutures fusion, multiple signaling pathways have been investigated to dissect the underlying mechanisms beneath the disease. In addition to genetic etiology, environment factors, especially mechanics, have also been proposed to have vital roles during the pathophysiological of craniosynostosis. However, the influence of mechanics factors in the cranial development remains largely unknown. In this review, we present a brief overview of the updated genetic mutations and environmental factors identified in both syndromic and nonsyndromic craniosynostosis. Furthermore, potential molecular signaling pathways and its relations have been described.
Collapse
Affiliation(s)
- Xiaowei Wu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, No. 22 Zhongguancun Avenue South, Haidian District, Beijing, 100081, PR. China
- National Engineering Laboratory for Digital and Material Technology of Stomatology,Beijing Key Laboratory of Digital Stomatology, No. 22 Zhongguancun Avenue South, Haidian District, Beijing, 100081, PR. China
| | - Yan Gu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, No. 22 Zhongguancun Avenue South, Haidian District, Beijing, 100081, PR. China
- National Engineering Laboratory for Digital and Material Technology of Stomatology,Beijing Key Laboratory of Digital Stomatology, No. 22 Zhongguancun Avenue South, Haidian District, Beijing, 100081, PR. China
| |
Collapse
|
37
|
Camp E, Pribadi C, Anderson PJ, Zannettino AC, Gronthos S. miRNA-376c-3p Mediates TWIST-1 Inhibition of Bone Marrow-Derived Stromal Cell Osteogenesis and Can Reduce Aberrant Bone Formation of TWIST-1 Haploinsufficient Calvarial Cells. Stem Cells Dev 2018; 27:1621-1633. [DOI: 10.1089/scd.2018.0083] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Esther Camp
- Mesenchymal Stem Cell Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Clara Pribadi
- Mesenchymal Stem Cell Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Peter J. Anderson
- Mesenchymal Stem Cell Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
- Australian Craniofacial Unit, Faculty of Health and Medical Sciences, Adelaide Medical School and Dentistry, Women's and Children's Hospital, The University of Adelaide, Adelaide, Australia
| | - Andrew C.W. Zannettino
- South Australian Health and Medical Research Institute, Adelaide, Australia
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
| |
Collapse
|
38
|
Teng CS, Ting MC, Farmer DT, Brockop M, Maxson RE, Crump JG. Altered bone growth dynamics prefigure craniosynostosis in a zebrafish model of Saethre-Chotzen syndrome. eLife 2018; 7:37024. [PMID: 30375332 PMCID: PMC6207424 DOI: 10.7554/elife.37024] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 10/14/2018] [Indexed: 01/09/2023] Open
Abstract
Cranial sutures separate the skull bones and house stem cells for bone growth and repair. In Saethre-Chotzen syndrome, mutations in TCF12 or TWIST1 ablate a specific suture, the coronal. This suture forms at a neural-crest/mesoderm interface in mammals and a mesoderm/mesoderm interface in zebrafish. Despite this difference, we show that combinatorial loss of TCF12 and TWIST1 homologs in zebrafish also results in specific loss of the coronal suture. Sequential bone staining reveals an initial, directional acceleration of bone production in the mutant skull, with subsequent localized stalling of bone growth prefiguring coronal suture loss. Mouse genetics further reveal requirements for Twist1 and Tcf12 in both the frontal and parietal bones for suture patency, and to maintain putative progenitors in the coronal region. These findings reveal conservation of coronal suture formation despite evolutionary shifts in embryonic origins, and suggest that the coronal suture might be especially susceptible to imbalances in progenitor maintenance and osteoblast differentiation. Some of the most common birth defects involve improper development of the head and face. One such birth defect is called craniosynostosis. Normally, an infant’s skull bones are not fully fused together. Instead, they are held together by soft tissue that allows the baby’s skull to more easily pass through the birth canal. This tissue also houses specialized cells called stem cells that allow the brain and skull to grow with the child. But in craniosynostosis these stem cells behave abnormally, which fuses the skull bones together and prevents the skull and brain from growing properly during childhood. One form of craniosynostosis called Saethre-Chotzen syndrome is caused by mutations in one of two genes that ensure the proper separation of two bones in the roof of the skull. Mice with mutations in the mouse versions of these genes develop the same problem and are used to study this condition. Mouse studies have looked mostly at what happens after birth. Studies looking at what happens in embryos with these mutations could help scientists learn more. One way to do so would be to genetically engineer zebrafish with the equivalent mutations. This is because zebrafish embryos are transparent and grow outside their mother’s body, making it easier for scientists to watch them develop. Now, Teng et al. have grown zebrafish with mutations in the zebrafish versions of the genes that cause Saethre-Chotzen syndrome. In the experiments, imaging tools were used to observe the live fish as they developed. This showed that the stem cells in their skulls become abnormal much earlier than previous studies had suggested. Teng et al. also showed that similar stem cells are responsible for growth of the skull in zebrafish and mice. Babies with craniosynostosis often need multiple, risky surgeries to separate their skull bones and allow their brain and head to grow. Unfortunately, these bones often fuse again because they have abnormal stem cells. Teng et al. provide new information on what goes wrong in these stem cells. Hopefully, this new information will help scientists to one day correct the defective stem cells in babies with craniosynostosis, thus reducing the number of surgeries needed to correct the problem.
Collapse
Affiliation(s)
- Camilla S Teng
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, United States.,Department of Biochemistry, Keck School of Medicine, University of Southern California, Los Angeles, United States
| | - Man-Chun Ting
- Department of Biochemistry, Keck School of Medicine, University of Southern California, Los Angeles, United States
| | - D'Juan T Farmer
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, United States
| | - Mia Brockop
- Department of Biochemistry, Keck School of Medicine, University of Southern California, Los Angeles, United States
| | - Robert E Maxson
- Department of Biochemistry, Keck School of Medicine, University of Southern California, Los Angeles, United States
| | - J Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, United States
| |
Collapse
|
39
|
Abstract
In 1993, Jabs et al. were the first to describe a genetic origin of craniosynostosis. Since this discovery, the genetic causes of the most common syndromes have been described. In 2015, a total of 57 human genes were reported for which there had been evidence that mutations were causally related to craniosynostosis. Facilitated by rapid technological developments, many others have been identified since then. Reviewing the literature, we characterize the most common craniosynostosis syndromes followed by a description of the novel causes that were identified between January 2015 and December 2017.
Collapse
Affiliation(s)
- Jacqueline A C Goos
- Department of Plastic and Reconstructive Surgery and Hand Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Irene M J Mathijssen
- Department of Plastic and Reconstructive Surgery and Hand Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
40
|
Parajuli P, Kumar S, Loumaye A, Singh P, Eragamreddy S, Nguyen TL, Ozkan S, Razzaque MS, Prunier C, Thissen JP, Atfi A. Twist1 Activation in Muscle Progenitor Cells Causes Muscle Loss Akin to Cancer Cachexia. Dev Cell 2018; 45:712-725.e6. [PMID: 29920276 PMCID: PMC6054474 DOI: 10.1016/j.devcel.2018.05.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 12/11/2017] [Accepted: 05/21/2018] [Indexed: 12/22/2022]
Abstract
Cancer cachexia is characterized by extreme skeletal muscle loss that results in high morbidity and mortality. The incidence of cachexia varies among tumor types, being lowest in sarcomas, whereas 90% of pancreatic ductal adenocarcinoma (PDAC) patients experience severe weight loss. How these tumors trigger muscle depletion is still unfolding. Serendipitously, we found that overexpression of Twist1 in mouse muscle progenitor cells, either constitutively during development or inducibly in adult animals, caused severe muscle atrophy with features reminiscent of cachexia. Using several genetic mouse models of PDAC, we detected a marked increase in Twist1 expression in muscle undergoing cachexia. In cancer patients, elevated levels of Twist1 are associated with greater degrees of muscle wasting. Finally, both genetic and pharmacological inactivation of Twist1 in muscle progenitor cells afforded substantial protection against cancer-mediated cachexia, which translated into meaningful survival benefits, implicating Twist1 as a possible target for attenuating muscle cachexia in cancer patients.
Collapse
Affiliation(s)
- Parash Parajuli
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Santosh Kumar
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Audrey Loumaye
- Endocrinology, Diabetology, and Nutrition Department, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Purba Singh
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Sailaja Eragamreddy
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Thien Ly Nguyen
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Seval Ozkan
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Mohammed S Razzaque
- Department of Applied Oral Sciences, The Forsyth Institute, Harvard School of Dental Medicine Affiliate, Cambridge, MA 02142, USA
| | - Céline Prunier
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris 75012, France
| | - Jean-Paul Thissen
- Endocrinology, Diabetology, and Nutrition Department, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Azeddine Atfi
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA; Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris 75012, France.
| |
Collapse
|
41
|
Mikheev AM, Mikheeva SA, Severs LJ, Funk CC, Huang L, McFaline-Figueroa JL, Schwensen J, Trapnell C, Price ND, Wong S, Rostomily RC. Targeting TWIST1 through loss of function inhibits tumorigenicity of human glioblastoma. Mol Oncol 2018; 12:1188-1202. [PMID: 29754406 PMCID: PMC6026950 DOI: 10.1002/1878-0261.12320] [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: 02/12/2018] [Revised: 04/16/2018] [Accepted: 04/25/2018] [Indexed: 12/30/2022] Open
Abstract
TWIST1 (TW) is a bHLH transcription factor (TF) and master regulator of the epithelial-to-mesenchymal transition (EMT). In vitro, TW promotes mesenchymal change, invasion, and self-renewal in glioblastoma (GBM) cells. However, the potential therapeutic relevance of TW has not been established through loss-of-function studies in human GBM cell xenograft models. The effects of TW loss of function (gene editing and knockdown) on inhibition of tumorigenicity of U87MG and GBM4 glioma stem cells were tested in orthotopic xenograft models and conditional knockdown in established flank xenograft tumors. RNAseq and the analysis of tumors investigated putative TW-associated mechanisms. Multiple bioinformatic tools revealed significant alteration of ECM, membrane receptors, signaling transduction kinases, and cytoskeleton dynamics leading to identification of PI3K/AKT signaling. We experimentally show alteration of AKT activity and periostin (POSTN) expression in vivo and/or in vitro. For the first time, we show that effect of TW knockout inhibits AKT activity in U87MG cells in vivo independent of PTEN mutation. The clinical relevance of TW and candidate mechanisms was established by analysis of the TCGA and ENCODE databases. TW expression was associated with decreased patient survival and LASSO regression analysis identified POSTN as one of top targets of TW in human GBM. While we previously demonstrated the role of TW in promoting EMT and invasion of glioma cells, these studies provide direct experimental evidence supporting protumorigenic role of TW independent of invasion in vivo and the therapeutic relevance of targeting TW in human GBM. Further, the role of TW driving POSTN expression and AKT signaling suggests actionable targets, which could be leveraged to mitigate the oncogenic effects of TW in GBM.
Collapse
Affiliation(s)
- Andrei M Mikheev
- Department of Neurosurgery, Houston Methodist Hospital and Research Institute, Houston, TX, USA.,Department of Neurosurgery and Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Svetlana A Mikheeva
- Department of Neurosurgery, Houston Methodist Hospital and Research Institute, Houston, TX, USA.,Department of Neurosurgery and Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Liza J Severs
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Cory C Funk
- Institute for Systems Biology, Seattle, WA, USA
| | - Lei Huang
- Department of Systems Medicine& Bioengineering, Houston Methodist Hospital and Research Institute, Weil Cornell Medical College, Houston, TX, USA
| | | | - Jeanette Schwensen
- Department of Neurosurgery and Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Cole Trapnell
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | | | - Stephen Wong
- Department of Systems Medicine& Bioengineering, Houston Methodist Hospital and Research Institute, Weil Cornell Medical College, Houston, TX, USA
| | - Robert C Rostomily
- Department of Neurosurgery, Houston Methodist Hospital and Research Institute, Houston, TX, USA.,Department of Neurosurgery and Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
42
|
Bai S, Li D, Xu L, Duan H, Yuan J, Wei M. Recombinant mouse periostin ameliorates coronal sutures fusion in Twist1 +/- mice. J Transl Med 2018; 16:103. [PMID: 29665811 PMCID: PMC5905175 DOI: 10.1186/s12967-018-1454-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 03/16/2018] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Saethre-Chotzen syndrome is an autosomal dominantly inherited disorder caused by mutations in the twist family basic helix-loop-helix transcription factor 1 (TWIST1) gene. Surgical procedures are frequently required to reduce morphological and functional defects in patients with Saethre-Chotzen syndrome. Therefore, the development of noninvasive procedures to treat Saethre-Chotzen syndrome is critical. We identified that periostin, which is an extracellular matrix protein that plays an important role in both bone and connective tissues, is downregulated in craniosynostosis patients. METHODS We aimed to verify the effects of different concentrations (0, 50, 100, and 200 μg/l) of recombinant mouse periostin in Twist1+/- mice (a mouse model of Saethre-Chotzen syndrome) coronal suture cells in vitro and in vivo. Cell proliferation, migration, and osteogenic differentiation were observed and detected. Twist1+/- mice were also injected with recombinant mouse periostin to verify the treatment effects. RESULTS Cell Counting Kit-8 results showed that recombinant mouse periostin inhibited the proliferation of suture-derived cells in a time- and concentration-dependent manner. Cell migration was also suppressed when treated with recombinant mouse periostin. Real-time quantitative PCR and Western blotting results suggested that messenger ribonucleic acid and protein expression of alkaline phosphatase, bone sialoprotein, collagen type I, and osteocalcin were all downregulated after treatment with recombinant mouse periostin. However, the expression of Wnt-3a, Wnt-1, and β-catenin were upregulated. The in vivo results demonstrated that periostin-treated Twist1+/- mice showed patent coronal sutures in comparison with non-treated Twist1+/- mice which have coronal craniosynostosis. CONCLUSION Our results suggest that recombinant mouse periostin can inhibit coronal suture cell proliferation and migration and suppress osteogenic differentiation of suture-derived cells via Wnt canonical signaling, as well as ameliorate coronal suture fusion in Twist1+/- mice.
Collapse
Affiliation(s)
- Shanshan Bai
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai, 200011, China
| | - Dong Li
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai, 200011, China
| | - Liang Xu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai, 200011, China
| | - Huichuan Duan
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai, 200011, China
| | - Jie Yuan
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai, 200011, China.
| | - Min Wei
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai, 200011, China.
| |
Collapse
|
43
|
Camp E, Anderson PJ, Zannettino ACW, Glackin CA, Gronthos S. Tyrosine kinase receptor c‐ros‐oncogene 1 inhibition alleviates aberrant bone formation of TWIST‐1 haploinsufficient calvarial cells from Saethre–Chotzen syndrome patients. J Cell Physiol 2018; 233:7320-7332. [DOI: 10.1002/jcp.26563] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 02/23/2018] [Indexed: 11/06/2022]
Affiliation(s)
- Esther Camp
- Mesenchymal Stem Cell LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Cancer ThemeSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
| | - Peter J. Anderson
- Cancer ThemeSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
- Australian Craniofacial UnitWomen's and Children's HospitalNorth AdelaideSouth AustraliaAustralia
| | - Andrew C. W. Zannettino
- Cancer ThemeSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
- Myeloma Research LaboratoryAdelaide Medical School, Faculty of Health and Medical SciencesUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | - Carlotta A. Glackin
- Molecular Medicine and NeurosciencesCity of Hope National Medical Center and Beckman Research InstituteDuarteCalifornia
| | - Stan Gronthos
- Mesenchymal Stem Cell LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Cancer ThemeSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
| |
Collapse
|
44
|
Apostolopoulou D, Kaxira OS, Hatzaki A, Panagopoulos KP, Alexandrou K, Stratoudakis A, Kollia P, Aleporou V. Genetic Analysis of Syndromic and Nonsyndromic Patients With Craniosynostosis Identifies Novel Mutations in the TWIST1 and EFNB1 Genes. Cleft Palate Craniofac J 2018; 55:1092-1102. [PMID: 29561715 DOI: 10.1177/1055665618760412] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Craniosynostosis, the premature fusion of cranial sutures, is usually divided into 2 major categories: syndromic and nonsyndromic. Mutations in the FGFR1, FGFR2, FGFR3, TWIST1, and EFNB1 genes cause the common craniosynostosis syndromes Muenke, Crouzon and Crouzon with acanthosis nigricans, Apert, Pfeiffer, Saethre-Chotzen, and Craniofrontonasal. Overlapping features among craniosynostosis syndromes, phenotypic heterogeneity even within the same syndrome, especially in the case of Muenke syndrome, and inadequate clinical evaluation can lead to misdiagnosis, which molecular testing can help clarify. OBJECTIVE The aim of this study is to investigate the underlying genetic cause in 46 patients with syndromic or nonsyndromic craniosynostosis by direct sequencing and/or microdeletion/microduplication analysis of the FGFR1-3, TWIST1, and EFNB1 genes. RESULTS Genetic analysis identified 3 novel mutations, c.413T>C - p.(Leu138Pro) [p.(L138P)] in TWIST1, the previously reported c.373G>A - p.(Glu125Lys) [p.(E125K)], and c.717dupA - p.(Leu240IlefsTer79) [p.(L240fs)] mutation in EFNB1 gene as well as 6 previously known mutations and a heterozygous TWIST1 gene deletion. The 2 novel mutations within EFNB1 gene arose de novo, but the novel mutation p.(L138P) within TWIST1 gene was inherited from the patient's father, who was found to be mosaic for the mutation. To our knowledge, this is the first case of mosaicism described for TWIST1 gene. CONCLUSIONS The contribution of molecular genetic analysis to the diagnosis of patients with syndromic craniosynostosis was useful because some were originally misdiagnosed. Conversely, thorough clinical evaluation can guide molecular testing and result in a correct diagnosis.
Collapse
Affiliation(s)
- Despina Apostolopoulou
- Department of Genetics and Biotechnology, School of Physical Sciences, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece.,AlfaLAB Molecular Biology and Cytogenetics Centre, Hygeia Group of Hospitals, Athens, Greece.,Hellenic Craniofacial Center, Athens, Greece
| | - Olga S Kaxira
- "MITERA" General, Maternity and Children Hospital, Athens, Greece
| | - Angeliki Hatzaki
- AlfaLAB Molecular Biology and Cytogenetics Centre, Hygeia Group of Hospitals, Athens, Greece
| | | | | | | | - Panagoula Kollia
- Department of Genetics and Biotechnology, School of Physical Sciences, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Vassiliki Aleporou
- Department of Genetics and Biotechnology, School of Physical Sciences, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
45
|
Twist1 regulates embryonic hematopoietic differentiation through binding to Myb and Gata2 promoter regions. Blood Adv 2017; 1:1672-1681. [PMID: 29296814 DOI: 10.1182/bloodadvances.2017006056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 07/26/2017] [Indexed: 02/07/2023] Open
Abstract
Mechanisms underlying differentiation of embryonic hematopoietic stem/progenitor cells (HSPCs) remain unclear. In mouse, intra-aortic clusters (IACs) form in the aorta-gonad-mesonephros region and acquire HSPC potential after 9.5 days postcoitum (dpc). In this study we demonstrate that Twist1 is highly expressed in c-Kit+CD31+CD34+ IACs, which are equivalent to embryonic HSPCs, compared with adult HSPCs. Progenitor activities of colony-forming unit (CFU) of granulocytes and macrophages, CFU of macrophages, burst-forming unit of erythroid, and B lymphopoiesis were impaired in IACs of Twist1-/- relative to wild-type embryos. Microarray analysis and real-time polymerase chain reaction showed downregulated expression of Myb and Gata2 transcripts in Twist1-/- IACs. Chromatin immunoprecipitation and promoter binding assays indicated that Twist1 directly binds the Myb and Gata2 promoters in 10.5-dpc IACs. We conclude that Twist1 is a novel transcriptional regulator of HSPC differentiation through direct binding to promoter regions of key regulators of the process.
Collapse
|
46
|
De novo mutations in inhibitors of Wnt, BMP, and Ras/ERK signaling pathways in non-syndromic midline craniosynostosis. Proc Natl Acad Sci U S A 2017; 114:E7341-E7347. [PMID: 28808027 DOI: 10.1073/pnas.1709255114] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Non-syndromic craniosynostosis (NSC) is a frequent congenital malformation in which one or more cranial sutures fuse prematurely. Mutations causing rare syndromic craniosynostoses in humans and engineered mouse models commonly increase signaling of the Wnt, bone morphogenetic protein (BMP), or Ras/ERK pathways, converging on shared nuclear targets that promote bone formation. In contrast, the genetics of NSC is largely unexplored. More than 95% of NSC is sporadic, suggesting a role for de novo mutations. Exome sequencing of 291 parent-offspring trios with midline NSC revealed 15 probands with heterozygous damaging de novo mutations in 12 negative regulators of Wnt, BMP, and Ras/ERK signaling (10.9-fold enrichment, P = 2.4 × 10-11). SMAD6 had 4 de novo and 14 transmitted mutations; no other gene had more than 1. Four familial NSC kindreds had mutations in genes previously implicated in syndromic disease. Collectively, these mutations contribute to 10% of probands. Mutations are predominantly loss-of-function, implicating haploinsufficiency as a frequent mechanism. A common risk variant near BMP2 increased the penetrance of SMAD6 mutations and was overtransmitted to patients with de novo mutations in other genes in these pathways, supporting a frequent two-locus pathogenesis. These findings implicate new genes in NSC and demonstrate related pathophysiology of common non-syndromic and rare syndromic craniosynostoses. These findings have implications for diagnosis, risk of recurrence, and risk of adverse neurodevelopmental outcomes. Finally, the use of pathways identified in rare syndromic disease to find genes accounting for non-syndromic cases may prove broadly relevant to understanding other congenital disorders featuring high locus heterogeneity.
Collapse
|
47
|
Bouard C, Terreux R, Tissier A, Jacqueroud L, Vigneron A, Ansieau S, Puisieux A, Payen L. Destabilization of the TWIST1/E12 complex dimerization following the R154P point-mutation of TWIST1: an in silico approach. BMC STRUCTURAL BIOLOGY 2017; 17:6. [PMID: 28521820 PMCID: PMC5437649 DOI: 10.1186/s12900-017-0076-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/02/2017] [Indexed: 11/24/2022]
Abstract
Background The bHLH transcription factor TWIST1 plays a key role in the embryonic development and in tumorigenesis. Some loss-of-function mutations of the TWIST1 gene have been shown to cause an autosomal dominant craniosynostosis, known as the Saethre-Chotzen syndrome (SCS). Although the functional impacts of many TWIST1 mutations have been experimentally reported, little is known on the molecular mechanisms underlying their loss-of-function. In a previous study, we highlighted the predictive value of in silico molecular dynamics (MD) simulations in deciphering the molecular function of TWIST1 residues. Results Here, since the substitution of the arginine 154 amino acid by a glycine residue (R154G) is responsible for the SCS phenotype and the substitution of arginine 154 by a proline experimentally decreases the dimerizing ability of TWIST1, we investigated the molecular impact of this point mutation using MD approaches. Consistently, MD simulations highlighted a clear decrease in the stability of the α-helix during the dimerization of the mutated R154P TWIST1/E12 dimer compared to the wild-type TE complex, which was further confirmed in vitro using immunoassays. Conclusions Our study demonstrates that MD simulations provide a structural explanation for the loss-of-function associated with the SCS TWIST1 mutation and provides a proof of concept of the predictive value of these MD simulations. This in silico methodology could be used to determine reliable pharmacophore sites, leading to the application of docking approaches in order to identify specific inhibitors of TWIST1 complexes. Electronic supplementary material The online version of this article (doi:10.1186/s12900-017-0076-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Charlotte Bouard
- Inserm UMR-S1052, Centre de Recherche en Cancérologie de Lyon, Lyon, 69373, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, 69373, France.,LabEX DEVweCAN, Lyon, France.,UNIV UMR1052, Lyon, 69008, France.,Centre Léon Bérard, Lyon, 69373, France
| | - Raphael Terreux
- Université de Lyon1, ISPB, Lyon, 69008, France.,Pole Rhône-Alpes de Bioinformatique - Lyon Gerland (PRABI-LG), Lyon, 69007, France.,CNRS UMR 5305, Lyon, France
| | - Agnès Tissier
- Inserm UMR-S1052, Centre de Recherche en Cancérologie de Lyon, Lyon, 69373, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, 69373, France.,LabEX DEVweCAN, Lyon, France.,UNIV UMR1052, Lyon, 69008, France.,Centre Léon Bérard, Lyon, 69373, France
| | - Laurent Jacqueroud
- Inserm UMR-S1052, Centre de Recherche en Cancérologie de Lyon, Lyon, 69373, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, 69373, France.,LabEX DEVweCAN, Lyon, France.,UNIV UMR1052, Lyon, 69008, France.,Centre Léon Bérard, Lyon, 69373, France
| | - Arnaud Vigneron
- Inserm UMR-S1052, Centre de Recherche en Cancérologie de Lyon, Lyon, 69373, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, 69373, France.,LabEX DEVweCAN, Lyon, France.,UNIV UMR1052, Lyon, 69008, France.,Centre Léon Bérard, Lyon, 69373, France
| | - Stéphane Ansieau
- Inserm UMR-S1052, Centre de Recherche en Cancérologie de Lyon, Lyon, 69373, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, 69373, France.,LabEX DEVweCAN, Lyon, France.,UNIV UMR1052, Lyon, 69008, France.,Centre Léon Bérard, Lyon, 69373, France
| | - Alain Puisieux
- Inserm UMR-S1052, Centre de Recherche en Cancérologie de Lyon, Lyon, 69373, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, 69373, France.,LabEX DEVweCAN, Lyon, France.,UNIV UMR1052, Lyon, 69008, France.,Centre Léon Bérard, Lyon, 69373, France.,Université de Lyon1, ISPB, Lyon, 69008, France.,Institut Universitaire de France, Paris, 75231, France
| | - Léa Payen
- Inserm UMR-S1052, Centre de Recherche en Cancérologie de Lyon, Lyon, 69373, France. .,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, 69373, France. .,LabEX DEVweCAN, Lyon, France. .,UNIV UMR1052, Lyon, 69008, France. .,Centre Léon Bérard, Lyon, 69373, France. .,Université de Lyon1, ISPB, Lyon, 69008, France. .,Laboratoire de Biochimie et Biologie Moléculaire (CHLS), Hospices Civils de Lyon, Lyon, 69003, France.
| |
Collapse
|
48
|
Jacqueroud L, Bouard C, Richard G, Payen L, Devouassoux-Shisheboran M, Spicer DB, Caramel J, Collin G, Puisieux A, Tissier A, Ansieau S. The Heterodimeric TWIST1-E12 Complex Drives the Oncogenic Potential of TWIST1 in Human Mammary Epithelial Cells. Neoplasia 2017; 18:317-327. [PMID: 27237323 PMCID: PMC4887617 DOI: 10.1016/j.neo.2016.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 03/18/2016] [Accepted: 03/28/2016] [Indexed: 11/26/2022] Open
Abstract
The TWIST1 embryonic transcription factor displays biphasic functions during the course of carcinogenesis. It facilitates the escape of cells from oncogene-induced fail-safe programs (senescence, apoptosis) and their consequent neoplastic transformation. Additionally, it promotes the epithelial-to-mesenchymal transition and the initiation of the metastatic spread of cancer cells. Interestingly, cancer cells recurrently remain dependent on TWIST1 for their survival and/or proliferation, making TWIST1 their Achilles’ heel. TWIST1 has been reported to form either homodimeric or heterodimeric complexes mainly in association with the E bHLH class I proteins. These complexes display distinct, sometimes even antagonistic, functions during development and unequal prometastatic functions in prostate cancer cells. Using a tethered dimer strategy, we successively assessed the ability of TWIST1 dimers to cooperate with an activated version of RAS in human mammary epithelial cell transformation, to provide mice with the ability to spontaneously develop breast tumors, and lastly to maintain a senescence program at a latent state in several breast cancer cell lines. We demonstrate that the TWIST1-E12 complex, unlike the homodimer, is an oncogenic form of TWIST1 in mammary epithelial cells and that efficient binding of both partners is a prerequisite for its activity. The detection of the heterodimer in human premalignant lesions by a proximity ligation assay, at a stage preceding the initiation of the metastatic cascade, is coherent with such an oncogenic function. TWIST1-E protein heterodimeric complexes may thus constitute the main active forms of TWIST1 with regard to senescence inhibition over the time course of breast tumorigenesis.
Collapse
Affiliation(s)
- Laurent Jacqueroud
- Inserm UMR-S1052, Centre de Recherche en Cancérologie de Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, France; LabEX DEVweCAN, Lyon, France; UNIV UMR1052, Lyon, France; Centre Léon Bérard, Lyon, France
| | - Charlotte Bouard
- Inserm UMR-S1052, Centre de Recherche en Cancérologie de Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, France; LabEX DEVweCAN, Lyon, France; UNIV UMR1052, Lyon, France; Centre Léon Bérard, Lyon, France
| | - Geoffrey Richard
- Inserm UMR-S1052, Centre de Recherche en Cancérologie de Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, France; LabEX DEVweCAN, Lyon, France; UNIV UMR1052, Lyon, France; Centre Léon Bérard, Lyon, France
| | - Léa Payen
- Inserm UMR-S1052, Centre de Recherche en Cancérologie de Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, France; LabEX DEVweCAN, Lyon, France; UNIV UMR1052, Lyon, France; Centre Léon Bérard, Lyon, France; Université de Lyon 1, ISPB, Lyon, France; Hospices Civils de Lyon, Lyon, France
| | - Mojgan Devouassoux-Shisheboran
- Inserm UMR-S1052, Centre de Recherche en Cancérologie de Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, France; LabEX DEVweCAN, Lyon, France; UNIV UMR1052, Lyon, France; Centre Léon Bérard, Lyon, France; Université de Lyon 1, ISPB, Lyon, France; Hospices Civils de Lyon, Lyon, France; Hôpital de la Croix-Rousse, Lyon, France
| | - Douglas B Spicer
- Center for Molecular Medicine, Main Medical Center Research Institute, Scarborough, ME, USA
| | - Julie Caramel
- Inserm UMR-S1052, Centre de Recherche en Cancérologie de Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, France; LabEX DEVweCAN, Lyon, France; UNIV UMR1052, Lyon, France; Centre Léon Bérard, Lyon, France; Université de Lyon 1, ISPB, Lyon, France
| | - Guillaume Collin
- Inserm UMR-S1052, Centre de Recherche en Cancérologie de Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, France; LabEX DEVweCAN, Lyon, France; UNIV UMR1052, Lyon, France; Centre Léon Bérard, Lyon, France
| | - Alain Puisieux
- Inserm UMR-S1052, Centre de Recherche en Cancérologie de Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, France; LabEX DEVweCAN, Lyon, France; UNIV UMR1052, Lyon, France; Centre Léon Bérard, Lyon, France; Université de Lyon 1, ISPB, Lyon, France; Institut Universitaire de France, Paris, France
| | - Agnès Tissier
- Inserm UMR-S1052, Centre de Recherche en Cancérologie de Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, France; LabEX DEVweCAN, Lyon, France; UNIV UMR1052, Lyon, France; Centre Léon Bérard, Lyon, France
| | - Stéphane Ansieau
- Inserm UMR-S1052, Centre de Recherche en Cancérologie de Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, France; LabEX DEVweCAN, Lyon, France; UNIV UMR1052, Lyon, France; Centre Léon Bérard, Lyon, France.
| |
Collapse
|
49
|
Forghanifard MM, Rad A, Farshchian M, Khaleghizadeh M, Gholamin M, Moghbeli M, Abbaszadegan MR. TWIST1 upregulates the MAGEA4 oncogene. Mol Carcinog 2017; 56:877-885. [DOI: 10.1002/mc.22541] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
| | - Abolfazl Rad
- Cellular and Molecular Research Center; Sabzevar University of Medical Sciences; Sabzevar Iran
| | - Moein Farshchian
- Molecular Medicine Research Department; ACECR-Khorasan Razavi Branch; Mashhad Iran
| | - Maryam Khaleghizadeh
- Division of Human Genetics; Immunology Research Center; Avicenna Research Institute; Mashhad University of Medical Sciences; Mashhad Iran
| | - Mehran Gholamin
- Division of Human Genetics; Immunology Research Center; Avicenna Research Institute; Mashhad University of Medical Sciences; Mashhad Iran
| | - Meysam Moghbeli
- Division of Human Genetics; Immunology Research Center; Avicenna Research Institute; Mashhad University of Medical Sciences; Mashhad Iran
| | - Mohammad Reza Abbaszadegan
- Division of Human Genetics; Immunology Research Center; Avicenna Research Institute; Mashhad University of Medical Sciences; Mashhad Iran
| |
Collapse
|
50
|
Lattanzi W, Barba M, Di Pietro L, Boyadjiev SA. Genetic advances in craniosynostosis. Am J Med Genet A 2017; 173:1406-1429. [PMID: 28160402 DOI: 10.1002/ajmg.a.38159] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 12/30/2016] [Accepted: 01/06/2017] [Indexed: 12/22/2022]
Abstract
Craniosynostosis, the premature ossification of one or more skull sutures, is a clinically and genetically heterogeneous congenital anomaly affecting approximately one in 2,500 live births. In most cases, it occurs as an isolated congenital anomaly, that is, nonsyndromic craniosynostosis (NCS), the genetic, and environmental causes of which remain largely unknown. Recent data suggest that, at least some of the midline NCS cases may be explained by two loci inheritance. In approximately 25-30% of patients, craniosynostosis presents as a feature of a genetic syndrome due to chromosomal defects or mutations in genes within interconnected signaling pathways. The aim of this review is to provide a detailed and comprehensive update on the genetic and environmental factors associated with NCS, integrating the scientific findings achieved during the last decade. Focus on the neurodevelopmental, imaging, and treatment aspects of NCS is also provided.
Collapse
Affiliation(s)
- Wanda Lattanzi
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Latium Musculoskeletal Tıssue Bank, Rome, Italy
| | - Marta Barba
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lorena Di Pietro
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Simeon A Boyadjiev
- Division of Genomic Medicine, Department of Pediatrics, Davis Medical Center, University of California, Sacramento, California
| |
Collapse
|