1
|
Ou W, Qi Z, Liu N, Zhang J, Mi X, Song Y, Fang Y, Cui B, Hou J, Yuan Z. Elucidating the role of TWIST1 in ulcerative colitis: a comprehensive bioinformatics and machine learning approach. Front Genet 2024; 15:1296570. [PMID: 38510272 PMCID: PMC10952112 DOI: 10.3389/fgene.2024.1296570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 02/16/2024] [Indexed: 03/22/2024] Open
Abstract
Background: Ulcerative colitis (UC) is a common and progressive inflammatory bowel disease primarily affecting the colon and rectum. Prolonged inflammation can lead to colitis-associated colorectal cancer (CAC). While the exact cause of UC remains unknown, this study aims to investigate the role of the TWIST1 gene in UC. Methods: Second-generation sequencing data from adult UC patients were obtained from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were identified, and characteristic genes were selected using machine learning and Lasso regression. The Receiver Operating Characteristic (ROC) curve assessed TWIST1's potential as a diagnostic factor (AUC score). Enriched pathways were analyzed, including Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Variation Analysis (GSVA). Functional mechanisms of marker genes were predicted, considering immune cell infiltration and the competing endogenous RNA (ceRNA) network. Results: We found 530 DEGs, with 341 upregulated and 189 downregulated genes. TWIST1 emerged as one of four potential UC biomarkers via machine learning. TWIST1 expression significantly differed in two datasets, GSE193677 and GSE83687, suggesting its diagnostic potential (AUC = 0.717 in GSE193677, AUC = 0.897 in GSE83687). Enrichment analysis indicated DEGs associated with TWIST1 were involved in processes like leukocyte migration, humoral immune response, and cell chemotaxis. Immune cell infiltration analysis revealed higher rates of M0 macrophages and resting NK cells in the high TWIST1 expression group, while TWIST1 expression correlated positively with M2 macrophages and resting NK cell infiltration. We constructed a ceRNA regulatory network involving 1 mRNA, 7 miRNAs, and 32 long non-coding RNAs (lncRNAs) to explore TWIST1's regulatory mechanism. Conclusion: TWIST1 plays a significant role in UC and has potential as a diagnostic marker. This study sheds light on UC's molecular mechanisms and underscores TWIST1's importance in its progression. Further research is needed to validate these findings in diverse populations and investigate TWIST1 as a therapeutic target in UC.
Collapse
Affiliation(s)
- Wenjie Ou
- School of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Zhaoxue Qi
- Department of Secretory Metabolism, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ning Liu
- General Surgery of The First Clinical Hospital of Jilin Academy of Chinese Medicine Sciences, Changchun, Jilin, China
| | - Junzi Zhang
- School of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xuguang Mi
- Department of Central Laboratory, Jilin Provincial People’s Hospital, Changchun, Jilin, China
| | - Yuan Song
- Department of Gastroenterology, Jilin Provincial People’s Hospital, Changchun, Jilin, China
| | - Yanqiu Fang
- Department of Central Laboratory, Jilin Provincial People’s Hospital, Changchun, Jilin, China
| | - Baiying Cui
- School of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Junjie Hou
- Department of Comprehensive Oncology, Jilin Provincial People’s Hospital, Changchun, Jilin, China
| | - Zhixin Yuan
- Department of Emergency Surgery, Jilin Provincial People’s Hospital, Changchun, Jilin, China
| |
Collapse
|
2
|
Balasubramanian D, Borges Pinto P, Grasso A, Vincent S, Tarayre H, Lajoignie D, Ghavi-Helm Y. Enhancer-promoter interactions can form independently of genomic distance and be functional across TAD boundaries. Nucleic Acids Res 2024; 52:1702-1719. [PMID: 38084924 PMCID: PMC10899756 DOI: 10.1093/nar/gkad1183] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 11/21/2023] [Accepted: 11/29/2023] [Indexed: 02/29/2024] Open
Abstract
Topologically Associating Domains (TADs) have been suggested to facilitate and constrain enhancer-promoter interactions. However, the role of TAD boundaries in effectively restricting these interactions remains unclear. Here, we show that a significant proportion of enhancer-promoter interactions are established across TAD boundaries in Drosophila embryos, but that developmental genes are strikingly enriched in intra- but not inter-TAD interactions. We pursued this observation using the twist locus, a master regulator of mesoderm development, and systematically relocated one of its enhancers to various genomic locations. While this developmental gene can establish inter-TAD interactions with its enhancer, the functionality of these interactions remains limited, highlighting the existence of topological constraints. Furthermore, contrary to intra-TAD interactions, the formation of inter-TAD enhancer-promoter interactions is not solely driven by genomic distance, with distal interactions sometimes favored over proximal ones. These observations suggest that other general mechanisms must exist to establish and maintain specific enhancer-promoter interactions across large distances.
Collapse
Affiliation(s)
- Deevitha Balasubramanian
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique UMR5242, Université Claude Bernard-Lyon 1; 69364 Lyon, France
- Indian Institute of Science Education and Research (IISER) Tirupati; Tirupati 517507 Andhra Pradesh, India
| | - Pedro Borges Pinto
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique UMR5242, Université Claude Bernard-Lyon 1; 69364 Lyon, France
| | - Alexia Grasso
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique UMR5242, Université Claude Bernard-Lyon 1; 69364 Lyon, France
| | - Séverine Vincent
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique UMR5242, Université Claude Bernard-Lyon 1; 69364 Lyon, France
| | - Hélène Tarayre
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique UMR5242, Université Claude Bernard-Lyon 1; 69364 Lyon, France
| | - Damien Lajoignie
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique UMR5242, Université Claude Bernard-Lyon 1; 69364 Lyon, France
| | - Yad Ghavi-Helm
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique UMR5242, Université Claude Bernard-Lyon 1; 69364 Lyon, France
| |
Collapse
|
3
|
Thiery JP, Sheng G, Shu X, Runyan R. How studies in developmental epithelial-mesenchymal transition and mesenchymal-epithelial transition inspired new research paradigms in biomedicine. Development 2024; 151:dev200128. [PMID: 38300897 DOI: 10.1242/dev.200128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Epithelial-mesenchymal transition (EMT) and its reverse mechanism, mesenchymal-epithelial transition (MET), are evolutionarily conserved mechanisms initially identified in studies of early metazoan development. EMT may even have been established in choanoflagellates, the closest unicellular relative of Metazoa. These crucial morphological transitions operate during body plan formation and subsequently in organogenesis. These findings have prompted an increasing number of investigators in biomedicine to assess the importance of such mechanisms that drive epithelial cell plasticity in multiple diseases associated with congenital disabilities and fibrosis, and, most importantly, in the progression of carcinoma. EMT and MET also play crucial roles in regenerative medicine, notably by contributing epigenetic changes in somatic cells to initiate reprogramming into stem cells and their subsequent differentiation into distinct lineages.
Collapse
Affiliation(s)
| | - Guojun Sheng
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Xiaodong Shu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Raymond Runyan
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
4
|
Huang L, Xing Y, Ning X, Yu Z, Bai X, Liu L, Sun S. Roles of Twist1 in lipid and glucose metabolism. Cell Commun Signal 2023; 21:270. [PMID: 37784111 PMCID: PMC10544448 DOI: 10.1186/s12964-023-01262-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/09/2023] [Indexed: 10/04/2023] Open
Abstract
The abnormal lipid and glucose metabolisms are linked to the metabolic disorders, tumorigenesis, and fibrotic diseases, which attracts the increasing attention to find out the key molecules involved in the lipid and glucose metabolism as the possible therapeutic targets on these diseases. A transcriptional factor Twist1 has been associated with not only the embryonic development, cancer, and fibrotic diseases, but also the regulation of lipid and glucose metabolism. In this review, we will discuss the roles and mechanisms of Twist1 in the obesity-associated white adipose tissue inflammation and insulin resistance, brown adipose tissue metabolism, fatty acid oxidation, and glucose metabolism in skeletal muscle to provide a rational perspective to consider Twist1 as a potential treatment target in clinic. Video Abstract.
Collapse
Affiliation(s)
- Liuyifei Huang
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Changle Road, No. 127 Changle West Road, Xi'an, Shaanxi, China
| | - Yan Xing
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Changle Road, No. 127 Changle West Road, Xi'an, Shaanxi, China
| | - Xiaoxuan Ning
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Changle Road, No. 127 Changle West Road, Xi'an, Shaanxi, China
| | - Zhixiang Yu
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Changle Road, No. 127 Changle West Road, Xi'an, Shaanxi, China
| | - Xiao Bai
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Changle Road, No. 127 Changle West Road, Xi'an, Shaanxi, China
| | - Limin Liu
- School of Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710032, Shaanxi, China.
| | - Shiren Sun
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Changle Road, No. 127 Changle West Road, Xi'an, Shaanxi, China.
| |
Collapse
|
5
|
Haerinck J, Goossens S, Berx G. The epithelial-mesenchymal plasticity landscape: principles of design and mechanisms of regulation. Nat Rev Genet 2023; 24:590-609. [PMID: 37169858 DOI: 10.1038/s41576-023-00601-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2023] [Indexed: 05/13/2023]
Abstract
Epithelial-mesenchymal plasticity (EMP) enables cells to interconvert between several states across the epithelial-mesenchymal landscape, thereby acquiring hybrid epithelial/mesenchymal phenotypic features. This plasticity is crucial for embryonic development and wound healing, but also underlies the acquisition of several malignant traits during cancer progression. Recent research using systems biology and single-cell profiling methods has provided novel insights into the main forces that shape EMP, which include the microenvironment, lineage specification and cell identity, and the genome. Additionally, key roles have emerged for hysteresis (cell memory) and cellular noise, which can drive stochastic transitions between cell states. Here, we review these forces and the distinct but interwoven layers of regulatory control that stabilize EMP states or facilitate epithelial-mesenchymal transitions (EMTs) and discuss the therapeutic potential of manipulating the EMP landscape.
Collapse
Affiliation(s)
- Jef Haerinck
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Steven Goossens
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Unit for Translational Research in Oncology, Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Geert Berx
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| |
Collapse
|
6
|
Gruss MJ, O’Callaghan C, Donnellan M, Corsi AK. A Twist-Box domain of the C. elegans Twist homolog, HLH-8, plays a complex role in transcriptional regulation. Genetics 2023; 224:iyad066. [PMID: 37067863 PMCID: PMC10411555 DOI: 10.1093/genetics/iyad066] [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: 11/04/2022] [Revised: 11/04/2022] [Accepted: 03/21/2023] [Indexed: 04/18/2023] Open
Abstract
TWIST1 is a basic helix-loop-helix (bHLH) transcription factor in humans that functions in mesoderm differentiation. TWIST1 primarily regulates genes as a transcriptional repressor often through TWIST-Box domain-mediated protein-protein interactions. The TWIST-Box also can function as an activation domain requiring 3 conserved, equidistant amino acids (LXXXFXXXR). Autosomal dominant mutations in TWIST1, including 2 reported in these conserved amino acids (F187L and R191M), lead to craniofacial defects in Saethre-Chotzen syndrome (SCS). Caenorhabditis elegans has a single TWIST1 homolog, HLH-8, that functions in the differentiation of the muscles responsible for egg laying and defecation. Null alleles in hlh-8 lead to severely egg-laying defective and constipated animals due to defects in the corresponding muscles. TWIST1 and HLH-8 share sequence identity in their bHLH regions; however, the domain responsible for the transcriptional activity of HLH-8 is unknown. Sequence alignment suggests that HLH-8 has a TWIST-Box LXXXFXXXR motif; however, its function also is unknown. CRISPR/Cas9 genome editing was utilized to generate a domain deletion and several missense mutations, including those analogous to SCS patients, in the 3 conserved HLH-8 amino acids to investigate their functional role. The TWIST-Box alleles did not phenocopy hlh-8 null mutants. The strongest phenotype detected was a retentive (Ret) phenotype with late-stage embryos in the hermaphrodite uterus. Further, GFP reporters of HLH-8 downstream target genes (arg-1::gfp and egl-15::gfp) revealed tissue-specific, target-specific, and allele-specific defects. Overall, the TWIST-Box in HLH-8 is partially required for the protein's transcriptional activity, and the conserved amino acids contribute unequally to the domain's function.
Collapse
Affiliation(s)
- Michael J Gruss
- Department of Biology, The Catholic University of America, 620 Michigan Ave., NE, Washington, D.C. 20064USA
| | - Colleen O’Callaghan
- Department of Biology, The Catholic University of America, 620 Michigan Ave., NE, Washington, D.C. 20064USA
| | - Molly Donnellan
- Department of Biology, The Catholic University of America, 620 Michigan Ave., NE, Washington, D.C. 20064USA
| | - Ann K Corsi
- Department of Biology, The Catholic University of America, 620 Michigan Ave., NE, Washington, D.C. 20064USA
| |
Collapse
|
7
|
Radhakrishnan K, Truong L, Carmichael CL. An "unexpected" role for EMT transcription factors in hematological development and malignancy. Front Immunol 2023; 14:1207360. [PMID: 37600794 PMCID: PMC10435889 DOI: 10.3389/fimmu.2023.1207360] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/14/2023] [Indexed: 08/22/2023] Open
Abstract
The epithelial to mesenchymal transition (EMT) is a fundamental developmental process essential for normal embryonic development. It is also important during various pathogenic processes including fibrosis, wound healing and epithelial cancer cell metastasis and invasion. EMT is regulated by a variety of cell signalling pathways, cell-cell interactions and microenvironmental cues, however the key drivers of EMT are transcription factors of the ZEB, TWIST and SNAIL families. Recently, novel and unexpected roles for these EMT transcription factors (EMT-TFs) during normal blood cell development have emerged, which appear to be largely independent of classical EMT processes. Furthermore, EMT-TFs have also begun to be implicated in the development and pathogenesis of malignant hematological diseases such as leukemia and lymphoma, and now present themselves or the pathways they regulate as possible new therapeutic targets within these malignancies. In this review, we discuss the ZEB, TWIST and SNAIL families of EMT-TFs, focusing on what is known about their normal roles during hematopoiesis as well as the emerging and "unexpected" contribution they play during development and progression of blood cancers.
Collapse
Affiliation(s)
- Karthika Radhakrishnan
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Lynda Truong
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Catherine L. Carmichael
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Monash University, Faculty of Medicine, Nursing and Health Sciences, Clayton, VIC, Australia
| |
Collapse
|
8
|
Toriumi K, Onodera Y, Takehara T, Mori T, Hasei J, Shigi K, Iwawaki N, Ozaki T, Akagi M, Nakanishi M, Teramura T. LRRC15 expression indicates high level of stemness regulated by TWIST1 in mesenchymal stem cells. iScience 2023; 26:106946. [PMID: 37534184 PMCID: PMC10391581 DOI: 10.1016/j.isci.2023.106946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 04/09/2023] [Accepted: 05/19/2023] [Indexed: 08/04/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are used as a major source for cell therapy, and its application is expanding in various diseases. On the other hand, reliable method to evaluate quality and therapeutic properties of MSC is limited. In this study, we focused on TWIST1 that is a transcription factor regulating stemness of MSCs and found that the transmembrane protein LRRC15 tightly correlated with the expression of TWIST1 and useful to expect TWIST1-regulated stemness of MSCs. The LRRC15-positive MSC populations in human and mouse bone marrow tissues highly expressed stemness-associated transcription factors and therapeutic cytokines, and showed better therapeutic effect in bleomycin-induced pulmonary fibrosis model mice. This study provides evidence for the important role of TWIST1 in the MSC stemness, and for the utility of the LRRC15 protein as a marker to estimate stem cell quality in MSCs before cell transplantation.
Collapse
Affiliation(s)
- Kensuke Toriumi
- Department of Orthopedic Surgery, Kindai University Faculty of Medicine, Osaka-sayama, Osaka, Japan
| | - Yuta Onodera
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Osaka-sayama, Osaka, Japan
| | - Toshiyuki Takehara
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Osaka-sayama, Osaka, Japan
| | - Tatsufumi Mori
- Life Science Institute, Kindai University, Osaka-sayama, Osaka, Japan
| | - Joe Hasei
- Department of Orthopedic Surgery, Okayama University Faculty of Medicine, Okayama, Okayama, Japan
| | - Kanae Shigi
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Osaka-sayama, Osaka, Japan
| | - Natsumi Iwawaki
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Osaka-sayama, Osaka, Japan
| | - Toshifumi Ozaki
- Department of Orthopedic Surgery, Okayama University Faculty of Medicine, Okayama, Okayama, Japan
| | - Masao Akagi
- Department of Orthopedic Surgery, Kindai University Faculty of Medicine, Osaka-sayama, Osaka, Japan
| | | | - Takeshi Teramura
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Osaka-sayama, Osaka, Japan
| |
Collapse
|
9
|
Sakaguchi S, Mizuno S, Okochi Y, Tanegashima C, Nishimura O, Uemura T, Kadota M, Naoki H, Kondo T. Single-cell transcriptome atlas of Drosophila gastrula 2.0. Cell Rep 2023:112707. [PMID: 37433294 DOI: 10.1016/j.celrep.2023.112707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/27/2023] [Accepted: 06/13/2023] [Indexed: 07/13/2023] Open
Abstract
During development, positional information directs cells to specific fates, leading them to differentiate with their own transcriptomes and express specific behaviors and functions. However, the mechanisms underlying these processes in a genome-wide view remain ambiguous, partly because the single-cell transcriptomic data of early developing embryos containing accurate spatial and lineage information are still lacking. Here, we report a single-cell transcriptome atlas of Drosophila gastrulae, divided into 77 transcriptomically distinct clusters. We find that the expression profiles of plasma-membrane-related genes, but not those of transcription-factor genes, represent each germ layer, supporting the nonequivalent contribution of each transcription-factor mRNA level to effector gene expression profiles at the transcriptome level. We also reconstruct the spatial expression patterns of all genes at the single-cell stripe level as the smallest unit. This atlas is an important resource for the genome-wide understanding of the mechanisms by which genes cooperatively orchestrate Drosophila gastrulation.
Collapse
Affiliation(s)
- Shunta Sakaguchi
- Laboratory of Cell Recognition and Pattern Formation, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Sonoko Mizuno
- Laboratory of Cell Recognition and Pattern Formation, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yasushi Okochi
- Laboratory of Theoretical Biology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan; Faculty of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Chiharu Tanegashima
- Laboratory for Phyloinformatics, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Osamu Nishimura
- Laboratory for Phyloinformatics, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Tadashi Uemura
- Laboratory of Cell Recognition and Pattern Formation, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan; Center for Living Systems Information Science, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Mitsutaka Kadota
- Laboratory for Phyloinformatics, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Honda Naoki
- Laboratory of Theoretical Biology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan; Laboratory of Data-driven Biology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashihiroshima, Hiroshima 739-8511, Japan; Theoretical Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan
| | - Takefumi Kondo
- Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan; The Keihanshin Consortium for Fostering the Next Generation of Global Leaders in Research (K-CONNEX), Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
10
|
You Y, Grasso E, Alvero A, Condon J, Dimova T, Hu A, Ding J, Alexandrova M, Manchorova D, Dimitrova V, Liao A, Mor G. Twist1-IRF9 Interaction Is Necessary for IFN-Stimulated Gene Anti-Zika Viral Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1899-1912. [PMID: 37144865 PMCID: PMC10615665 DOI: 10.4049/jimmunol.2300081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/10/2023] [Indexed: 05/06/2023]
Abstract
An efficient immune defense against pathogens requires sufficient basal sensing mechanisms that can deliver prompt responses. Type I IFNs are protective against acute viral infections and respond to viral and bacterial infections, but their efficacy depends on constitutive basal activity that promotes the expression of downstream genes known as IFN-stimulated genes (ISGs). Type I IFNs and ISGs are constitutively produced at low quantities and yet exert profound effects essential for numerous physiological processes beyond antiviral and antimicrobial defense, including immunomodulation, cell cycle regulation, cell survival, and cell differentiation. Although the canonical response pathway for type I IFNs has been extensively characterized, less is known regarding the transcriptional regulation of constitutive ISG expression. Zika virus (ZIKV) infection is a major risk for human pregnancy complications and fetal development and depends on an appropriate IFN-β response. However, it is poorly understood how ZIKV, despite an IFN-β response, causes miscarriages. We have uncovered a mechanism for this function specifically in the context of the early antiviral response. Our results demonstrate that IFN regulatory factor (IRF9) is critical in the early response to ZIKV infection in human trophoblast. This function is contingent on IRF9 binding to Twist1. In this signaling cascade, Twist1 was not only a required partner that promotes IRF9 binding to the IFN-stimulated response element but also an upstream regulator that controls basal levels of IRF9. The absence of Twist1 renders human trophoblast cells susceptible to ZIKV infection.
Collapse
Affiliation(s)
- Yuan You
- C. S Mott Center for Human Development, Wayne State University, 275 E Hancock St, Detroit, MI, 48093
| | - Esteban Grasso
- C. S Mott Center for Human Development, Wayne State University, 275 E Hancock St, Detroit, MI, 48093
- School of Science, University of Buenos Aires, Intendente Guiraldes 2160, Buenos Aires, 1428
| | - Ayesha Alvero
- C. S Mott Center for Human Development, Wayne State University, 275 E Hancock St, Detroit, MI, 48093
| | - Jennifer Condon
- C. S Mott Center for Human Development, Wayne State University, 275 E Hancock St, Detroit, MI, 48093
| | - Tanya Dimova
- Institute of Biology and Immunology of Reproduction “Acad. K. Bratanov”, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Anna Hu
- C. S Mott Center for Human Development, Wayne State University, 275 E Hancock St, Detroit, MI, 48093
| | - Jiahui Ding
- C. S Mott Center for Human Development, Wayne State University, 275 E Hancock St, Detroit, MI, 48093
| | - Marina Alexandrova
- Institute of Biology and Immunology of Reproduction “Acad. K. Bratanov”, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Diana Manchorova
- Institute of Biology and Immunology of Reproduction “Acad. K. Bratanov”, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Violeta Dimitrova
- Institute of Biology and Immunology of Reproduction “Acad. K. Bratanov”, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Aihua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Gil Mor
- C. S Mott Center for Human Development, Wayne State University, 275 E Hancock St, Detroit, MI, 48093
| |
Collapse
|
11
|
Rose M, Domsch K, Bartle-Schultheis J, Reim I, Schaub C. Twist regulates Yorkie activity to guide lineage reprogramming of syncytial alary muscles. Cell Rep 2022; 38:110295. [DOI: 10.1016/j.celrep.2022.110295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/25/2021] [Accepted: 01/04/2022] [Indexed: 11/28/2022] Open
|
12
|
Mishra N, Heisenberg CP. Dissecting Organismal Morphogenesis by Bridging Genetics and Biophysics. Annu Rev Genet 2021; 55:209-233. [PMID: 34460295 DOI: 10.1146/annurev-genet-071819-103748] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Multicellular organisms develop complex shapes from much simpler, single-celled zygotes through a process commonly called morphogenesis. Morphogenesis involves an interplay between several factors, ranging from the gene regulatory networks determining cell fate and differentiation to the mechanical processes underlying cell and tissue shape changes. Thus, the study of morphogenesis has historically been based on multidisciplinary approaches at the interface of biology with physics and mathematics. Recent technological advances have further improved our ability to study morphogenesis by bridging the gap between the genetic and biophysical factors through the development of new tools for visualizing, analyzing, and perturbing these factors and their biochemical intermediaries. Here, we review how a combination of genetic, microscopic, biophysical, and biochemical approaches has aided our attempts to understand morphogenesis and discuss potential approaches that may be beneficial to such an inquiry in the future. Expected final online publication date for the Annual Review of Genetics, Volume 55 is November 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Nikhil Mishra
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria; ,
| | | |
Collapse
|
13
|
Denk-Lobnig M, Totz JF, Heer NC, Dunkel J, Martin AC. Combinatorial patterns of graded RhoA activation and uniform F-actin depletion promote tissue curvature. Development 2021; 148:dev199232. [PMID: 34124762 PMCID: PMC8254875 DOI: 10.1242/dev.199232] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 05/04/2021] [Indexed: 01/03/2023]
Abstract
During development, gene expression regulates cell mechanics and shape to sculpt tissues. Epithelial folding proceeds through distinct cell shape changes that occur simultaneously in different regions of a tissue. Here, using quantitative imaging in Drosophila melanogaster, we investigate how patterned cell shape changes promote tissue bending during early embryogenesis. We find that the transcription factors Twist and Snail combinatorially regulate a multicellular pattern of lateral F-actin density that differs from the previously described Myosin-2 gradient. This F-actin pattern correlates with whether cells apically constrict, stretch or maintain their shape. We show that the Myosin-2 gradient and F-actin depletion do not depend on force transmission, suggesting that transcriptional activity is required to create these patterns. The Myosin-2 gradient width results from a gradient in RhoA activation that is refined through the balance between RhoGEF2 and the RhoGAP C-GAP. Our experimental results and simulations of a 3D elastic shell model show that tuning gradient width regulates tissue curvature.
Collapse
Affiliation(s)
- Marlis Denk-Lobnig
- Biology Department, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Jan F. Totz
- Mathematics Department, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Natalie C. Heer
- Biology Department, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Jörn Dunkel
- Mathematics Department, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Adam C. Martin
- Biology Department, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| |
Collapse
|
14
|
The role of epithelial-mesenchymal transition-regulating transcription factors in anti-cancer drug resistance. Arch Pharm Res 2021; 44:281-292. [PMID: 33768509 PMCID: PMC8009775 DOI: 10.1007/s12272-021-01321-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 03/14/2021] [Indexed: 12/16/2022]
Abstract
The complex orchestration of gene expression that mediates the transition of epithelial cells into mesenchymal cells is implicated in cancer development and metastasis. As the primary regulator of the process, epithelial-mesenchymal transition-regulating transcription factors (EMT-TFs) play key roles in metastasis. They are also highlighted in recent preclinical studies on resistance to cancer therapy. This review describes the role of three main EMT-TFs, including Snail, Twist1, and zinc-finger E homeobox-binding 1 (ZEB1), relating to drug resistance and current possible approaches for future challenges targeting EMT-TFs.
Collapse
|
15
|
Plygawko AT, Kan S, Campbell K. Epithelial-mesenchymal plasticity: emerging parallels between tissue morphogenesis and cancer metastasis. Philos Trans R Soc Lond B Biol Sci 2020; 375:20200087. [PMID: 32829692 PMCID: PMC7482222 DOI: 10.1098/rstb.2020.0087] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Many cells possess epithelial–mesenchymal plasticity (EMP), which allows them to shift reversibly between adherent, static and more detached, migratory states. These changes in cell behaviour are driven by the programmes of epithelial–mesenchymal transition (EMT) and mesenchymal–epithelial transition (MET), both of which play vital roles during normal development and tissue homeostasis. However, the aberrant activation of these processes can also drive distinct stages of cancer progression, including tumour invasiveness, cell dissemination and metastatic colonization and outgrowth. This review examines emerging common themes underlying EMP during tissue morphogenesis and malignant progression, such as the context dependence of EMT transcription factors, a central role for partial EMTs and the nonlinear relationship between EMT and MET. This article is part of a discussion meeting issue ‘Contemporary morphogenesis'.
Collapse
Affiliation(s)
- Andrew T Plygawko
- Department of Biomedical Science and Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
| | - Shohei Kan
- Department of Biomedical Science and Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
| | - Kyra Campbell
- Department of Biomedical Science and Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
16
|
Gheisari E, Aakhte M, Müller HAJ. Gastrulation in Drosophila melanogaster: Genetic control, cellular basis and biomechanics. Mech Dev 2020; 163:103629. [PMID: 32615151 DOI: 10.1016/j.mod.2020.103629] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 06/08/2020] [Accepted: 06/24/2020] [Indexed: 01/31/2023]
Abstract
Gastrulation is generally understood as the morphogenetic processes that result in the spatial organization of the blastomere into the three germ layers, ectoderm, mesoderm and endoderm. This review summarizes our current knowledge of the morphogenetic mechanisms in Drosophila gastrulation. In addition to the events that drive mesoderm invagination and germband elongation, we pay particular attention to other, less well-known mechanisms including midgut invagination, cephalic furrow formation, dorsal fold formation, and mesoderm layer formation. This review covers topics ranging from the identification and functional characterization of developmental and morphogenetic control genes to the analysis of the physical properties of cells and tissues and the control of cell and tissue mechanics of the morphogenetic movements in the gastrula.
Collapse
Affiliation(s)
- Elham Gheisari
- Institute for Biology, Dept. Developmental Genetics, University of Kassel, Germany
| | - Mostafa Aakhte
- Institute for Biology, Dept. Developmental Genetics, University of Kassel, Germany
| | - H-Arno J Müller
- Institute for Biology, Dept. Developmental Genetics, University of Kassel, Germany.
| |
Collapse
|
17
|
Yang J, Antin P, Berx G, Blanpain C, Brabletz T, Bronner M, Campbell K, Cano A, Casanova J, Christofori G, Dedhar S, Derynck R, Ford HL, Fuxe J, García de Herreros A, Goodall GJ, Hadjantonakis AK, Huang RYJ, Kalcheim C, Kalluri R, Kang Y, Khew-Goodall Y, Levine H, Liu J, Longmore GD, Mani SA, Massagué J, Mayor R, McClay D, Mostov KE, Newgreen DF, Nieto MA, Puisieux A, Runyan R, Savagner P, Stanger B, Stemmler MP, Takahashi Y, Takeichi M, Theveneau E, Thiery JP, Thompson EW, Weinberg RA, Williams ED, Xing J, Zhou BP, Sheng G. Guidelines and definitions for research on epithelial-mesenchymal transition. Nat Rev Mol Cell Biol 2020; 21:341-352. [PMID: 32300252 PMCID: PMC7250738 DOI: 10.1038/s41580-020-0237-9] [Citation(s) in RCA: 1222] [Impact Index Per Article: 244.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2020] [Indexed: 02/06/2023]
Abstract
Epithelial–mesenchymal transition (EMT) encompasses dynamic changes in cellular organization from epithelial to mesenchymal phenotypes, which leads to functional changes in cell migration and invasion. EMT occurs in a diverse range of physiological and pathological conditions and is driven by a conserved set of inducing signals, transcriptional regulators and downstream effectors. With over 5,700 publications indexed by Web of Science in 2019 alone, research on EMT is expanding rapidly. This growing interest warrants the need for a consensus among researchers when referring to and undertaking research on EMT. This Consensus Statement, mediated by ‘the EMT International Association’ (TEMTIA), is the outcome of a 2-year-long discussion among EMT researchers and aims to both clarify the nomenclature and provide definitions and guidelines for EMT research in future publications. We trust that these guidelines will help to reduce misunderstanding and misinterpretation of research data generated in various experimental models and to promote cross-disciplinary collaboration to identify and address key open questions in this research field. While recognizing the importance of maintaining diversity in experimental approaches and conceptual frameworks, we emphasize that lasting contributions of EMT research to increasing our understanding of developmental processes and combatting cancer and other diseases depend on the adoption of a unified terminology to describe EMT. In this Consensus Statement, the authors (on behalf of the EMT International Association) propose guidelines to define epithelial–mesenchymal transition, its phenotypic plasticity and the associated multiple intermediate epithelial–mesenchymal cell states. Clarification of nomenclature and definitions will help reduce misinterpretation of research data generated in different experimental model systems and promote cross-disciplinary collaboration.
Collapse
Affiliation(s)
- Jing Yang
- Departments of Pharmacology and Pediatrics, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.
| | - Parker Antin
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Geert Berx
- Molecular and Cellular Oncology Lab, Department of Biomedical Molecular Biology, Ghent University, Cancer Research Institute Ghent (CRIG), VIB Center for Inflammation Research, Ghent, Belgium
| | - Cédric Blanpain
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Marianne Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Kyra Campbell
- Department of Biomedical Science and Bateson Centre, University of Sheffield, Sheffield, UK
| | - Amparo Cano
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), IdiPAZ & Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Jordi Casanova
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology/Institut de Biologia Molecular de Barcelona (IBMB-CSIC), Barcelona, Spain
| | | | - Shoukat Dedhar
- Department of Biochemistry and Molecular Biology, University of British Columbia and British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Rik Derynck
- Departments of Cell and Tissue Biology, and Anatomy, University of California at San Francisco, San Francisco, CA, USA
| | - Heide L Ford
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jonas Fuxe
- Department of Laboratory Medicine (LABMED), Division of Pathology, Karolinska University Hospital and Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Antonio García de Herreros
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM) and Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - Gregory J Goodall
- Centre for Cancer Biology, An alliance of SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ruby Y J Huang
- School of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chaya Kalcheim
- Department of Medical Neurobiology, Institute for medical Research Israel-Canada and the Safra Center for Neurosciences, Hebrew University of Jerusalem, Hadassah Medical School, Jerusalem, Israel
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, MD Anderson Cancer Center, Houston, TX, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Yeesim Khew-Goodall
- Centre for Cancer Biology, an Alliance of SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Herbert Levine
- Department of Physics, Northeastern University, Boston, MA, USA
| | - Jinsong Liu
- Department of Anatomic Pathology, The Division of Pathology and Laboratory Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gregory D Longmore
- Department of Medicine (Oncology) and Department of Cell Biology and Physiology, ICCE Institute, Washington University, St. Louis, MO, USA
| | - Sendurai A Mani
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joan Massagué
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, London, UK
| | - David McClay
- Department of Biology, Duke University, Durham, NC, USA
| | - Keith E Mostov
- Departments of Anatomy and Biochemistry/Biophysics, University of California, San Francisco, School of Medicine, San Francisco, CA, USA
| | - Donald F Newgreen
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia
| | - M Angela Nieto
- Instituto de Neurociencias (CSIC-UMH) Avda Ramon y Cajal s/n, Sant Joan d´Alacant, Spain
| | - Alain Puisieux
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France.,Institut Curie, PSL Research University, Paris, France
| | - Raymond Runyan
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Pierre Savagner
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, University Paris-Saclay, Villejuif, France
| | - Ben Stanger
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marc P Stemmler
- Department of Experimental Medicine 1, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Yoshiko Takahashi
- Department of Zoology, Graduate School of Science, Kyoto University, Kyoto, Japan
| | | | - Eric Theveneau
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Jean Paul Thiery
- Guangzhou Regenerative Medicine and Health, Guangdong Laboratory, Guangzhou, China
| | - Erik W Thompson
- School of Biomedical Sciences and Institute of Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Robert A Weinberg
- Whitehead Institute for Biomedical Research, Department of Biology, MIT Ludwig Center for Molecular Oncology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Elizabeth D Williams
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q) and Queensland Bladder Cancer Initiative (QBCI), School of Biomedical Sciences and Institute of Health and Biomedical Innovation, Queensland University of Technology, Woolloongabba, QLD, Australia
| | - Jianhua Xing
- Department of Computational and Systems Biology and UPMC-Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Binhua P Zhou
- Department of Molecular and Cellular Biochemistry and UK Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Guojun Sheng
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan.
| | | |
Collapse
|
18
|
Mahmoud M, Souilhol C, Serbanovic-Canic J, Evans P. GATA4-Twist1 Signalling in Disturbed Flow-Induced Atherosclerosis. Cardiovasc Drugs Ther 2020; 33:231-237. [PMID: 30809744 DOI: 10.1007/s10557-019-06863-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Endothelial cell (EC) dysfunction (enhanced inflammation, proliferation and permeability) is the initial trigger for atherosclerosis. Atherosclerosis shows preferential development near branches and bends exposed to disturbed blood flow. By contrast, sites that are exposed to non-disturbed blood flow are atheroprotected. Disturbed flow promotes atherosclerosis by promoting EC dysfunction. Blood flow controls EC function through transcriptional and post-transcriptional mechanisms that are incompletely understood. METHODS AND RESULTS We identified the developmental transcription factors Twist1 and GATA4 as being enriched in EC at disturbed flow, atheroprone regions of the porcine aorta in a microarray study. Further work using the porcine and murine aortae demonstrated that Twist1 and GATA4 expression was enhanced at the atheroprone, disturbed flow sites in vivo. Using controlled in vitro flow systems, the expression of Twist1 and GATA4 was enhanced under disturbed compared to non-disturbed flow in cultured cells. Disturbed flow promoted Twist1 expression through a GATA4-mediated transcriptional mechanism as revealed by a series of in vivo and in vitro studies. GATA4-Twist1 signalling promoted EC proliferation, inflammation, permeability and endothelial-to-mesenchymal transition (EndoMT) under disturbed flow, leading to atherosclerosis development, as shown in a combination of in vitro and in vivo studies using GATA4 and Twist1-specific siRNA and EC-specific GATA4 and Twist1 Knock out (KO) mice. CONCLUSIONS We revealed that GATA4-Twist1-Snail signalling triggers EC dysfunction and atherosclerosis; this work could lead to the development of novel anti-atherosclerosis therapeutics.
Collapse
Affiliation(s)
- Marwa Mahmoud
- Department of Biomedical Engineering, The City College of New York, New York, NY, 10031, USA.
| | - Celine Souilhol
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, UK
| | - Jovana Serbanovic-Canic
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, UK
| | - Paul Evans
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, UK
| |
Collapse
|
19
|
Bandodkar PU, Al Asafen H, Reeves GT. Spatiotemporal control of gene expression boundaries using a feedforward loop. Dev Dyn 2020; 249:369-382. [PMID: 31925874 DOI: 10.1002/dvdy.150] [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: 10/11/2019] [Revised: 12/24/2019] [Accepted: 01/02/2020] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND A feedforward loop (FFL) is commonly observed in several biological networks. The FFL network motif has been mostly studied with respect to variation of the input signal in time, with only a few studies of FFL activity in a spatially distributed system such as morphogen-mediated tissue patterning. However, most morphogen gradients also evolve in time. RESULTS We studied the spatiotemporal behavior of a coherent FFL in two contexts: (a) a generic, oscillating morphogen gradient and (b) the dorsal-ventral patterning of the early Drosophila embryo by a gradient of the NF-κB homolog dorsal with its early target Twist. In both models, we found features in the dynamics of the intermediate node-phase difference and noise filtering-that were largely independent of the parameterization of the models, and thus were functions of the structure of the FFL itself. In the dorsal gradient model, we also found that proper target gene expression was not possible without including the effect of maternal pioneer factor Zelda. CONCLUSIONS An FFL buffers fluctuation to changes in the morphogen signal ensuring stable gene expression boundaries.
Collapse
Affiliation(s)
- Prasad U Bandodkar
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina
| | - Hadel Al Asafen
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina
| | - Gregory T Reeves
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
20
|
Ji LJ, Su J, Xu AL, Pang B, Huang QM. MiR-134-5p attenuates neuropathic pain progression through targeting Twist1. J Cell Biochem 2019; 120:1694-1701. [PMID: 30187947 DOI: 10.1002/jcb.27486] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/18/2018] [Indexed: 01/24/2023]
Abstract
Neuropathic pain is a kind of chronic pain because of dysfunctions of somatosensory nerve system. Recently, many studies have demonstrated that microRNAs (miRs) play crucial roles in neuropathic pain development. This study was designed to investigate the effects of miR-134-5p on the process of neuropathic pain progression in a rat model established by chronic sciatic nerve injury (CCI). First, we observed that miR-134-5p was significantly decreased in CCI rat models. Overexpression of miR-134-5p strongly alleviated neuropathic pain behaviors including mechanical and thermal hyperalgesia. Meanwhile, inflammatory cytokine expression, such as IL-6, IL-1β and TNF-α in CCI rats were greatly repressed by upregulation of miR-134-5p. Twist1 has been widely regarded as a poor prognosis biomarker in diverse diseases. Here, by using bioinformatic analysis, 3'-untranslated region (UTR) of Twist1 was predicted to be a downstream target of miR-134-5p in our study. Here, we found that overexpression of miR-134-5p was able to suppress Twist1 dramatically. Furthermore, it was exhibited that Twist1 was increased in CCI rats time-dependently and Twist1 was inhibited in vivo. Subsequently, downregulation of Twist1 in CCI rats could depress neuropathic pain progression via inhibiting neuroinflammation. In conclusion, our current study indicated that miR-134-5p may inhibit neuropathic pain development through targeting Twist1. Our findings suggested that miR-134-5p might provide a novel therapeutic target for neuropathic pain.
Collapse
Affiliation(s)
- Li-Juan Ji
- Department of Sport Medicine and Rehabilitation Center, School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jing Su
- Department of Rehabilitation, The Affiliated Huai'an Hospital of Xuzhou Medical College and The Second People's Hospital of Huai'an, Huai'an, China
| | - An-Le Xu
- Department of Sport Medicine and Rehabilitation Center, School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Bo Pang
- Department of Sport Medicine and Rehabilitation Center, School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Qiang-Min Huang
- Department of Sport Medicine and Rehabilitation Center, School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
21
|
Tan M, Asad M, Heong V, Wong MK, Tan TZ, Ye J, Kuay KT, Thiery JP, Scott C, Huang RYJ. The FZD7-TWIST1 axis is responsible for anoikis resistance and tumorigenesis in ovarian carcinoma. Mol Oncol 2019; 13:757-780. [PMID: 30548372 PMCID: PMC6441896 DOI: 10.1002/1878-0261.12425] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/24/2018] [Accepted: 11/20/2018] [Indexed: 12/20/2022] Open
Abstract
Frizzled family receptor 7 (FZD7), a Wnt signaling receptor, is associated with the maintenance of stem cell properties and cancer progression. FZD7 has emerged as a potential therapeutic target because it is capable of transducing both canonical and noncanonical Wnt signals. In this study, we investigated the regulatory pathway downstream of FZD7 and its functional roles. We found that FZD7 expression was crucial to the maintenance of the mesenchymal phenotype, anoikis resistance, and spheroid and tumor formation in ovarian cancer (OC). We identified TWIST1 as the crucial downstream effector of the FZD7 pathway. TWIST1, a basic helix loop helix transcription factor, is known to associate with mesenchymal and cancer stem cell phenotypes. Manipulating TWIST1 expression mimicked the functional consequences observed in the FZD7 model, and overexpression of TWIST1 partially rescued the functional phenotypes abolished by FZD7 knockdown. We further proved that FZD7 regulated TWIST1 expression through epigenetic modifications of H3K4me3 and H3K27ac at the TWIST1 proximal promoter. We also identified that the FZD7‐TWIST1 axis regulates the expression of BCL2, a gene that controls apoptosis. Identification of this FZD7‐TWIST1‐BCL2 pathway reaffirms the mechanism of anoikis resistance in OC. We subsequently showed that the FZD7‐TWIST1 axis can be targeted by using a small molecule inhibitor of porcupine, an enzyme essential for secretion and functional activation of Wnts. In conclusion, our results identified that the FZD7‐TWIST1 axis is important for tumorigenesis and anoikis resistance, and therapeutic inhibition results in cell death in OCs.
Collapse
Affiliation(s)
- Ming Tan
- Cancer Science Institute of Singapore, Singapore.,Center for Translational Medicine, National University of Singapore, Singapore
| | - Mohammad Asad
- Cancer Science Institute of Singapore, Singapore.,Center for Translational Medicine, National University of Singapore, Singapore.,Department of Obstetrics and Gynaecology, National University Hospital of Singapore, Singapore
| | - Valerie Heong
- Cancer Science Institute of Singapore, Singapore.,Center for Translational Medicine, National University of Singapore, Singapore.,Department of Haematology-Oncology, National University Cancer Institute Singapore, Singapore.,Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Meng Kang Wong
- Cancer Science Institute of Singapore, Singapore.,Center for Translational Medicine, National University of Singapore, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, Singapore.,Center for Translational Medicine, National University of Singapore, Singapore
| | - Jieru Ye
- Cancer Science Institute of Singapore, Singapore.,Center for Translational Medicine, National University of Singapore, Singapore
| | - Kuee Theng Kuay
- Cancer Science Institute of Singapore, Singapore.,Center for Translational Medicine, National University of Singapore, Singapore
| | - Jean Paul Thiery
- Cancer Science Institute of Singapore, Singapore.,Center for Translational Medicine, National University of Singapore, Singapore.,Department of Biochemistry, National University of Singapore, Singapore.,Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| | - Clare Scott
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Ruby Yun-Ju Huang
- Cancer Science Institute of Singapore, Singapore.,Center for Translational Medicine, National University of Singapore, Singapore.,Department of Obstetrics and Gynaecology, National University Hospital of Singapore, Singapore.,Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
22
|
Takenouchi T, Sakamoto Y, Sato H, Suzuki H, Uehara T, Ohsone Y, Kosaki K. Ablepharon and craniosynostosis in a patient with a localized TWIST1 basic domain substitution. Am J Med Genet A 2018; 176:2777-2780. [PMID: 30450715 DOI: 10.1002/ajmg.a.40525] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/20/2018] [Accepted: 08/02/2018] [Indexed: 11/11/2022]
Abstract
The TWIST family is a group of highly conserved basic helix-loop-helix transcription factors. In humans, TWIST1 haploinsufficiency causes Saethre-Chotzen syndrome, which is characterized by craniosynostosis. Heterozygous localized TWIST1 and TWIST2 basic domain substitutions exert antimorphic effects to cause Sweeney-Cox syndrome, Barber-Say syndrome, and ablepharon-macrostomia syndrome, respectively. Sweeney-Cox syndrome, Barber-Say syndrome, and ablepharon-macrostomia syndrome share the facial features of ablepharon, hypertelorism, underdevelopment of the eyelids, and cheek pads adjacent to the corners of the mouth. Existence of phenotypic overlap between Saethre-Chotzen syndrome and Sweeney-Cox syndrome remains unknown. Herein, we document a male infant with the distinctive facial features of ablepharon, hypertelorism, cheek pads adjacent to the corners of the mouth, and bilateral coronal suture craniosynostosis who had a de novo heterozygous mutation in the basic domain of TWIST1, that is, c.351C>G p.Glu117Asp. The pathogenicity of this variant was supported by in silico and in vivo evidence. Our review showed that Sweeney-Cox syndrome appears to share many characteristics with Barber-Say syndrome and ablepharon-macrostomia syndrome except for craniosynostosis, which is a cardinal feature of Saethre-Chotzen syndrome. An amino acid substitution from Glu117 to Asp, both of which are the sole members of negatively charged amino acids, resulted in a prototypic Sweeney-Cox syndrome phenotype. This suggests that any amino acid substitutions at Glu117 would likely lead to the Sweeney-Cox syndrome phenotype or lethality. The present observation suggests that a localized TWIST1 basic domain substitution, that is, p.Glu117Asp, in TWIST1 may exert a mild antimorphic effect similar to that of haploinsufficiency, leading to craniosynostosis and ablepharon.
Collapse
Affiliation(s)
- Toshiki Takenouchi
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Yoshiaki Sakamoto
- Plastic and Reconstructive Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hironori Sato
- Department of Pediatrics, Chiba University, Chiba City, Japan
| | - Hisato Suzuki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Tomoko Uehara
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | | | - Kenjiro Kosaki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
23
|
Quarto N, Shailendra S, Meyer NP, Menon S, Renda A, Longaker MT. Twist1-Haploinsufficiency Selectively Enhances the Osteoskeletal Capacity of Mesoderm-Derived Parietal Bone Through Downregulation of Fgf23. Front Physiol 2018; 9:1426. [PMID: 30374308 PMCID: PMC6196243 DOI: 10.3389/fphys.2018.01426] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 09/19/2018] [Indexed: 02/05/2023] Open
Abstract
Craniofacial development is a program exquisitely orchestrated by tissue contributions and regulation of genes expression. The basic helix–loop–helix (bHLH) transcription factor Twist1 expressed in the skeletal mesenchyme is a key regulator of craniofacial development playing an important role during osteoskeletogenesis. This study investigates the postnatal impact of Twist1 haploinsufficiency on the osteoskeletal ability and regeneration on two calvarial bones arising from tissues of different embryonic origin: the neural crest-derived frontal and the mesoderm-derived parietal bones. We show that Twist1 haplonsufficiency as well Twist1-sh-mediated silencing selectively enhanced osteogenic and tissue regeneration ability of mesoderm-derived bones. Transcriptomic profiling, gain-and loss-of-function experiments revealed that Twist1 haplonsufficiency triggers its selective activity on mesoderm-derived bone through a sharp downregulation of the bone-derived hormone Fgf23 that is upregulated exclusively in wild-type parietal bone.
Collapse
Affiliation(s)
- Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, CA, United States.,Dipartimento di Scienze Biomediche Avanzate, Universita' degli Studi di Napoli Federico II, Naples, Italy
| | - Siny Shailendra
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, CA, United States
| | - Nathaniel P Meyer
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, CA, United States
| | - Siddharth Menon
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, CA, United States
| | - Andrea Renda
- Dipartimento di Scienze Biomediche Avanzate, Universita' degli Studi di Napoli Federico II, Naples, Italy
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, CA, United States
| |
Collapse
|
24
|
Kasai Y, Stahl S, Crews S. Specification of the Drosophila CNS midline cell lineage: direct control of single-minded transcription by dorsal/ventral patterning genes. Gene Expr 2018; 7:171-89. [PMID: 9840810 PMCID: PMC6151948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The Drosophila CNS consists of a bilaterally symmetric group of neurons separated by a discrete group of CNS midline cells. The specification of the CNS midline cell lineage requires transcription of the single-minded gene. Genetic evidence suggests that a group of transcription factors, including Dorsal, Snail, Twist, and Daughterless:: Scute, is required for initial single-minded transcription. Comparison of the DNA sequences of the single-minded gene regulatory regions between two Drosophila species reveals conserved sequence elements. Biochemical studies using purified proteins indicate that a number of these conserved sequences represent binding sites for Dorsal, Snail, and Twist. In vitro mutagenesis combined with germline transformation indicates that these binding sites are required in vivo for single-minded mesectodermal transcription. These results show that single-minded transcription and, thus, CNS midline specification is directly controlled by dorsal/ventral patterning transcription factors. They also suggest a model in which multiple transcriptional activators function in a cooperative, concentration-dependent mode in combination with a transcriptional repressor to restrict single-minded transcription to the CNS midline precursor cells.
Collapse
Affiliation(s)
- Yumi Kasai
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7260
| | - Stephanie Stahl
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7260
| | - Stephen Crews
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7260
- Address correspondence to Stephen Crews, Department of Biochemistry and Biophysics, Mary Ellen Jones Bldg., The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7260. Tel: (919) 962-4380; Fax: (919) 962-3155; E-mail:
| |
Collapse
|
25
|
Vincent-Mistiaen Z, Elbediwy A, Vanyai H, Cotton J, Stamp G, Nye E, Spencer-Dene B, Thomas GJ, Mao J, Thompson B. YAP drives cutaneous squamous cell carcinoma formation and progression. eLife 2018; 7:e33304. [PMID: 30231971 PMCID: PMC6147738 DOI: 10.7554/elife.33304] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 06/29/2018] [Indexed: 01/14/2023] Open
Abstract
Squamous cell carcinoma (SCC) can progress to malignant metastatic cancer, including an aggressive subtype known as spindle cell carcinoma (spSCC). spSCC formation involves epithelial-to-mesenchymal transition (EMT), yet the molecular basis of this event remains unknown. The transcriptional co-activator YAP undergoes recurrent amplification in human SCC and overexpression of YAP drives SCC formation in mice. Here, we show that human spSCC tumours also feature strong nuclear localisation of YAP and overexpression of activated YAP (NLS-YAP-5SA) with Keratin-5 (K5-CreERt) is sufficient to induce rapid formation of both SCC and spSCC in mice. spSCC tumours arise at sites of epithelial scratch wounding, where tumour-initiating epithelial cells undergo EMT to generate spSCC. Expression of the EMT transcription factor ZEB1 arises upon wounding and is a defining characteristic of spSCC in mice and humans. Thus, the wound healing response synergises with YAP to drive metaplastic transformation of SCC to spSCC.
Collapse
Affiliation(s)
| | | | | | - Jennifer Cotton
- University of Massachusetts Medical SchoolWorcesterUnited States
| | | | - Emma Nye
- The Francis Crick InstituteLondonUnited Kingdom
| | | | - Gareth J Thomas
- Cancer Sciences Unit, Faculty of MedicineUniversity of SouthamptonSouthamptonUnited Kingdom
| | - Junhao Mao
- University of Massachusetts Medical SchoolWorcesterUnited States
| | | |
Collapse
|
26
|
A novel fluorinated triazole derivative suppresses macrophage activation and alleviates experimental colitis via a Twist1-dependent pathway. Biochem Pharmacol 2018; 155:275-287. [DOI: 10.1016/j.bcp.2018.07.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/16/2018] [Indexed: 02/01/2023]
|
27
|
Chen SC, Liao TT, Yang MH. Emerging roles of epithelial-mesenchymal transition in hematological malignancies. J Biomed Sci 2018; 25:37. [PMID: 29685144 PMCID: PMC5913878 DOI: 10.1186/s12929-018-0440-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/19/2018] [Indexed: 01/06/2023] Open
Abstract
Background Epithelial-mesenchymal transition is an important process in embryonic development, fibrosis, and cancer metastasis. During the progression of epithelial cancer, activation of epithelial-mesenchymal transition is tightly associated with metastasis, stemness and drug resistance. However, the role of epithelial-mesenchymal transition in non-epithelial cancer is relatively unclear. Main body Epithelial-mesenchymal transition transcription factors are critical in both myeloid and lymphoid development. Growing evidence indicates their roles in cancer cells to promote leukemia and lymphoma progression. The expression of epithelial-mesenchymal transition transcription factors can cause the differentiation of indolent type to the aggressive type of lymphoma. Their up-regulation confers cancer cells resistant to chemotherapy, tyrosine kinase inhibitors, and radiotherapy. Conversely, the down-regulation of epithelial-mesenchymal transition transcription factors, monoclonal antibodies, induce lymphoma cells apoptosis. Conclusions Epithelial-mesenchymal transition transcription factors are potentially important prognostic or predictive factors and treatment targets for leukemia and lymphoma.
Collapse
Affiliation(s)
- San-Chi Chen
- Institute of Clinical Medicine, National Yang-Ming University, No. 155, Sec. 2, Li-Nong Street, Taipei, 11221, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tsai-Tsen Liao
- Institute of Clinical Medicine, National Yang-Ming University, No. 155, Sec. 2, Li-Nong Street, Taipei, 11221, Taiwan.,Cancer Progression Center of Excellence, National Yang-Ming University, Taipei, Taiwan.,Department of Otolaryngology, National Yang-Ming University, Taipei, Taiwan
| | - Muh-Hwa Yang
- Institute of Clinical Medicine, National Yang-Ming University, No. 155, Sec. 2, Li-Nong Street, Taipei, 11221, Taiwan. .,Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan. .,Cancer Progression Center of Excellence, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
28
|
Abstract
Craniosynostosis is the premature fusion of the calvarial sutures that is associated with a number of physical and intellectual disabilities spanning from pediatric to adult years. Over the past two decades, techniques in molecular genetics and more recently, advances in high-throughput DNA sequencing have been used to examine the underlying pathogenesis of this disease. To date, mutations in 57 genes have been identified as causing craniosynostosis and the number of newly discovered genes is growing rapidly as a result of the advances in genomic technologies. While contributions from both genetic and environmental factors in this disease are increasingly apparent, there remains a gap in knowledge that bridges the clinical characteristics and genetic markers of craniosynostosis with their signaling pathways and mechanotransduction processes. By linking genotype to phenotype, outlining the role of cell mechanics may further uncover the specific mechanotransduction pathways underlying craniosynostosis. Here, we present a brief overview of the recent findings in craniofacial genetics and cell mechanics, discussing how this information together with animal models is advancing our understanding of craniofacial development.
Collapse
Affiliation(s)
- Zeinab Al-Rekabi
- Department of Mechanical Engineering, University of Washington, 3900 E Stevens Way NE, Seattle, WA, 98195, USA
- Seattle Children’s Research Institute, Center for Developmental Biology and Regenerative Medicine, 1900 9 Ave, Seattle, WA, 98101, USA
| | - Michael L. Cunningham
- Seattle Children’s Research Institute, Center for Developmental Biology and Regenerative Medicine, 1900 9 Ave, Seattle, WA, 98101, USA
- Department of Pediatrics, Division of Craniofacial Medicine and the, University of Washington, 1959 NE Pacific St., Seattle, WA, 98195, USA
| | - Nathan J. Sniadecki
- Department of Mechanical Engineering, University of Washington, 3900 E Stevens Way NE, Seattle, WA, 98195, USA
- Department of Bioengineering, University of Washington, 3720 15 Ave NE, Seattle WA, 98105, USA
| |
Collapse
|
29
|
Kim JS, Jiménez BIM, Kwak HJ, Park SC, Xiao P, Weisblat DA, Cho SJ. Spatiotemporal expression of a twist homolog in the leech Helobdella austinensis. Dev Genes Evol 2017; 227:245-252. [PMID: 28699036 DOI: 10.1007/s00427-017-0585-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 06/27/2017] [Indexed: 11/28/2022]
Abstract
Genes of the twist family encode bHLH transcription factors known to be involved in the regulation and differentiation of early mesoderm. Here, we report our characterization of Hau-twist, a twist homolog from the leech Helobdella austinensis, a tractable lophotrochozoan representative. Hau-twist was expressed in segmental founder cells of the mesodermal lineage, in subsets of cells within the mesodermal lineage of the germinal plate, in circumferential muscle fibers of a provisional integument during segmentation and organogenesis stages and on the ventral side of the developing proboscis. Thus, consistent with other systems, our results suggest that twist gene of the leech Helobdella might function in mesoderm differentiation.
Collapse
Affiliation(s)
- Jin-Se Kim
- School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Brenda Irene Medina Jiménez
- School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Hee-Jin Kwak
- School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Soon Cheol Park
- Department of Life Sciences, Chung-Ang University, Seoul, 156-756, Republic of Korea
| | - Ping Xiao
- Department of Molecular and Cell Biology, 385 LSA, University of California, Berkeley, CA, 94720-3200, USA
| | - David A Weisblat
- Department of Molecular and Cell Biology, 385 LSA, University of California, Berkeley, CA, 94720-3200, USA.
| | - Sung-Jin Cho
- School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea. .,Department of Molecular and Cell Biology, 385 LSA, University of California, Berkeley, CA, 94720-3200, USA.
| |
Collapse
|
30
|
Wieschaus E, Nüsslein-Volhard C. The Heidelberg Screen for Pattern Mutants of Drosophila: A Personal Account. Annu Rev Cell Dev Biol 2016; 32:1-46. [PMID: 27501451 DOI: 10.1146/annurev-cellbio-113015-023138] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In large-scale mutagenesis screens performed in 1979-1980 at the EMBL in Heidelberg, we isolated mutations affecting the pattern or structure of the larval cuticle in Drosophila. The 600 mutants we characterized could be assigned to 120 genes and represent the majority of such genes in the genome. These mutants subsequently provided a rich resource for understanding many fundamental developmental processes, such as the transcriptional hierarchies controlling segmentation, the establishment of cell states by signaling pathways, and the differentiation of epithelial cells. Most of the Heidelberg genes are now molecularly known, and many of them are conserved in other animals, including humans. Although the screens were initially driven entirely by curiosity, the mutants now serve as models for many human diseases. In this review, we describe the rationale of the screening procedures and provide a classification of the genes on the basis of their initial phenotypes and the subsequent molecular analyses.
Collapse
Affiliation(s)
- Eric Wieschaus
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544;
| | | |
Collapse
|
31
|
Mahmoud MM, Kim HR, Xing R, Hsiao S, Mammoto A, Chen J, Serbanovic-Canic J, Feng S, Bowden NP, Maguire R, Ariaans M, Francis SE, Weinberg PD, van der Heiden K, Jones EA, Chico TJA, Ridger V, Evans PC. TWIST1 Integrates Endothelial Responses to Flow in Vascular Dysfunction and Atherosclerosis. Circ Res 2016; 119:450-62. [PMID: 27245171 PMCID: PMC4959828 DOI: 10.1161/circresaha.116.308870] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 05/20/2016] [Accepted: 05/27/2016] [Indexed: 11/18/2022]
Abstract
RATIONALE Blood flow-induced shear stress controls endothelial cell (EC) physiology during atherosclerosis via transcriptional mechanisms that are incompletely understood. The mechanosensitive transcription factor TWIST is expressed during embryogenesis, but its role in EC responses to shear stress and focal atherosclerosis is unknown. OBJECTIVE To investigate whether TWIST regulates endothelial responses to shear stress during vascular dysfunction and atherosclerosis and compare TWIST function in vascular development and disease. METHODS AND RESULTS The expression and function of TWIST1 was studied in EC in both developing vasculature and during the initiation of atherosclerosis. In zebrafish, twist was expressed in early embryonic vasculature where it promoted angiogenesis by inducing EC proliferation and migration. In adult porcine and murine arteries, TWIST1 was expressed preferentially at low shear stress regions as evidenced by quantitative polymerase chain reaction and en face staining. Moreover, studies of experimental murine carotid arteries and cultured EC revealed that TWIST1 was induced by low shear stress via a GATA4-dependent transcriptional mechanism. Gene silencing in cultured EC and EC-specific genetic deletion in mice demonstrated that TWIST1 promoted atherosclerosis by inducing inflammation and enhancing EC proliferation associated with vascular leakiness. CONCLUSIONS TWIST expression promotes developmental angiogenesis by inducing EC proliferation and migration. In addition to its role in development, TWIST is expressed preferentially at low shear stress regions of adult arteries where it promotes atherosclerosis by inducing EC proliferation and inflammation. Thus, pleiotropic functions of TWIST control vascular disease and development.
Collapse
Affiliation(s)
- Marwa M Mahmoud
- From the Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom (M.M.M., H.R.K., S.H., J.S.-C., S.F., N.P.B., R.M., M.A., S.E.F., T.J.A.C., V.R., P.C.E.); ERASMUS MC, Rotterdam, The Netherlands (R.X., K.v.d.H.); Vascular Biology Program, Department of Surgery (A.M.) and Department of Ophthalmology (J.C.), Boston Children's Hospital, Harvard Medical School, MA; Department of Bioengineering, Imperial College London, London, United Kingdom (P.D.W.); and Department of Cardiovascular Science, Katholieke Universiteit Leuven, Leuven, Belgium (E.A.J.)
| | - Hyejeong Rosemary Kim
- From the Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom (M.M.M., H.R.K., S.H., J.S.-C., S.F., N.P.B., R.M., M.A., S.E.F., T.J.A.C., V.R., P.C.E.); ERASMUS MC, Rotterdam, The Netherlands (R.X., K.v.d.H.); Vascular Biology Program, Department of Surgery (A.M.) and Department of Ophthalmology (J.C.), Boston Children's Hospital, Harvard Medical School, MA; Department of Bioengineering, Imperial College London, London, United Kingdom (P.D.W.); and Department of Cardiovascular Science, Katholieke Universiteit Leuven, Leuven, Belgium (E.A.J.)
| | - Rouyu Xing
- From the Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom (M.M.M., H.R.K., S.H., J.S.-C., S.F., N.P.B., R.M., M.A., S.E.F., T.J.A.C., V.R., P.C.E.); ERASMUS MC, Rotterdam, The Netherlands (R.X., K.v.d.H.); Vascular Biology Program, Department of Surgery (A.M.) and Department of Ophthalmology (J.C.), Boston Children's Hospital, Harvard Medical School, MA; Department of Bioengineering, Imperial College London, London, United Kingdom (P.D.W.); and Department of Cardiovascular Science, Katholieke Universiteit Leuven, Leuven, Belgium (E.A.J.)
| | - Sarah Hsiao
- From the Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom (M.M.M., H.R.K., S.H., J.S.-C., S.F., N.P.B., R.M., M.A., S.E.F., T.J.A.C., V.R., P.C.E.); ERASMUS MC, Rotterdam, The Netherlands (R.X., K.v.d.H.); Vascular Biology Program, Department of Surgery (A.M.) and Department of Ophthalmology (J.C.), Boston Children's Hospital, Harvard Medical School, MA; Department of Bioengineering, Imperial College London, London, United Kingdom (P.D.W.); and Department of Cardiovascular Science, Katholieke Universiteit Leuven, Leuven, Belgium (E.A.J.)
| | - Akiko Mammoto
- From the Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom (M.M.M., H.R.K., S.H., J.S.-C., S.F., N.P.B., R.M., M.A., S.E.F., T.J.A.C., V.R., P.C.E.); ERASMUS MC, Rotterdam, The Netherlands (R.X., K.v.d.H.); Vascular Biology Program, Department of Surgery (A.M.) and Department of Ophthalmology (J.C.), Boston Children's Hospital, Harvard Medical School, MA; Department of Bioengineering, Imperial College London, London, United Kingdom (P.D.W.); and Department of Cardiovascular Science, Katholieke Universiteit Leuven, Leuven, Belgium (E.A.J.)
| | - Jing Chen
- From the Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom (M.M.M., H.R.K., S.H., J.S.-C., S.F., N.P.B., R.M., M.A., S.E.F., T.J.A.C., V.R., P.C.E.); ERASMUS MC, Rotterdam, The Netherlands (R.X., K.v.d.H.); Vascular Biology Program, Department of Surgery (A.M.) and Department of Ophthalmology (J.C.), Boston Children's Hospital, Harvard Medical School, MA; Department of Bioengineering, Imperial College London, London, United Kingdom (P.D.W.); and Department of Cardiovascular Science, Katholieke Universiteit Leuven, Leuven, Belgium (E.A.J.)
| | - Jovana Serbanovic-Canic
- From the Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom (M.M.M., H.R.K., S.H., J.S.-C., S.F., N.P.B., R.M., M.A., S.E.F., T.J.A.C., V.R., P.C.E.); ERASMUS MC, Rotterdam, The Netherlands (R.X., K.v.d.H.); Vascular Biology Program, Department of Surgery (A.M.) and Department of Ophthalmology (J.C.), Boston Children's Hospital, Harvard Medical School, MA; Department of Bioengineering, Imperial College London, London, United Kingdom (P.D.W.); and Department of Cardiovascular Science, Katholieke Universiteit Leuven, Leuven, Belgium (E.A.J.)
| | - Shuang Feng
- From the Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom (M.M.M., H.R.K., S.H., J.S.-C., S.F., N.P.B., R.M., M.A., S.E.F., T.J.A.C., V.R., P.C.E.); ERASMUS MC, Rotterdam, The Netherlands (R.X., K.v.d.H.); Vascular Biology Program, Department of Surgery (A.M.) and Department of Ophthalmology (J.C.), Boston Children's Hospital, Harvard Medical School, MA; Department of Bioengineering, Imperial College London, London, United Kingdom (P.D.W.); and Department of Cardiovascular Science, Katholieke Universiteit Leuven, Leuven, Belgium (E.A.J.)
| | - Neil P Bowden
- From the Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom (M.M.M., H.R.K., S.H., J.S.-C., S.F., N.P.B., R.M., M.A., S.E.F., T.J.A.C., V.R., P.C.E.); ERASMUS MC, Rotterdam, The Netherlands (R.X., K.v.d.H.); Vascular Biology Program, Department of Surgery (A.M.) and Department of Ophthalmology (J.C.), Boston Children's Hospital, Harvard Medical School, MA; Department of Bioengineering, Imperial College London, London, United Kingdom (P.D.W.); and Department of Cardiovascular Science, Katholieke Universiteit Leuven, Leuven, Belgium (E.A.J.)
| | - Richard Maguire
- From the Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom (M.M.M., H.R.K., S.H., J.S.-C., S.F., N.P.B., R.M., M.A., S.E.F., T.J.A.C., V.R., P.C.E.); ERASMUS MC, Rotterdam, The Netherlands (R.X., K.v.d.H.); Vascular Biology Program, Department of Surgery (A.M.) and Department of Ophthalmology (J.C.), Boston Children's Hospital, Harvard Medical School, MA; Department of Bioengineering, Imperial College London, London, United Kingdom (P.D.W.); and Department of Cardiovascular Science, Katholieke Universiteit Leuven, Leuven, Belgium (E.A.J.)
| | - Markus Ariaans
- From the Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom (M.M.M., H.R.K., S.H., J.S.-C., S.F., N.P.B., R.M., M.A., S.E.F., T.J.A.C., V.R., P.C.E.); ERASMUS MC, Rotterdam, The Netherlands (R.X., K.v.d.H.); Vascular Biology Program, Department of Surgery (A.M.) and Department of Ophthalmology (J.C.), Boston Children's Hospital, Harvard Medical School, MA; Department of Bioengineering, Imperial College London, London, United Kingdom (P.D.W.); and Department of Cardiovascular Science, Katholieke Universiteit Leuven, Leuven, Belgium (E.A.J.)
| | - Sheila E Francis
- From the Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom (M.M.M., H.R.K., S.H., J.S.-C., S.F., N.P.B., R.M., M.A., S.E.F., T.J.A.C., V.R., P.C.E.); ERASMUS MC, Rotterdam, The Netherlands (R.X., K.v.d.H.); Vascular Biology Program, Department of Surgery (A.M.) and Department of Ophthalmology (J.C.), Boston Children's Hospital, Harvard Medical School, MA; Department of Bioengineering, Imperial College London, London, United Kingdom (P.D.W.); and Department of Cardiovascular Science, Katholieke Universiteit Leuven, Leuven, Belgium (E.A.J.)
| | - Peter D Weinberg
- From the Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom (M.M.M., H.R.K., S.H., J.S.-C., S.F., N.P.B., R.M., M.A., S.E.F., T.J.A.C., V.R., P.C.E.); ERASMUS MC, Rotterdam, The Netherlands (R.X., K.v.d.H.); Vascular Biology Program, Department of Surgery (A.M.) and Department of Ophthalmology (J.C.), Boston Children's Hospital, Harvard Medical School, MA; Department of Bioengineering, Imperial College London, London, United Kingdom (P.D.W.); and Department of Cardiovascular Science, Katholieke Universiteit Leuven, Leuven, Belgium (E.A.J.)
| | - Kim van der Heiden
- From the Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom (M.M.M., H.R.K., S.H., J.S.-C., S.F., N.P.B., R.M., M.A., S.E.F., T.J.A.C., V.R., P.C.E.); ERASMUS MC, Rotterdam, The Netherlands (R.X., K.v.d.H.); Vascular Biology Program, Department of Surgery (A.M.) and Department of Ophthalmology (J.C.), Boston Children's Hospital, Harvard Medical School, MA; Department of Bioengineering, Imperial College London, London, United Kingdom (P.D.W.); and Department of Cardiovascular Science, Katholieke Universiteit Leuven, Leuven, Belgium (E.A.J.)
| | - Elizabeth A Jones
- From the Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom (M.M.M., H.R.K., S.H., J.S.-C., S.F., N.P.B., R.M., M.A., S.E.F., T.J.A.C., V.R., P.C.E.); ERASMUS MC, Rotterdam, The Netherlands (R.X., K.v.d.H.); Vascular Biology Program, Department of Surgery (A.M.) and Department of Ophthalmology (J.C.), Boston Children's Hospital, Harvard Medical School, MA; Department of Bioengineering, Imperial College London, London, United Kingdom (P.D.W.); and Department of Cardiovascular Science, Katholieke Universiteit Leuven, Leuven, Belgium (E.A.J.)
| | - Timothy J A Chico
- From the Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom (M.M.M., H.R.K., S.H., J.S.-C., S.F., N.P.B., R.M., M.A., S.E.F., T.J.A.C., V.R., P.C.E.); ERASMUS MC, Rotterdam, The Netherlands (R.X., K.v.d.H.); Vascular Biology Program, Department of Surgery (A.M.) and Department of Ophthalmology (J.C.), Boston Children's Hospital, Harvard Medical School, MA; Department of Bioengineering, Imperial College London, London, United Kingdom (P.D.W.); and Department of Cardiovascular Science, Katholieke Universiteit Leuven, Leuven, Belgium (E.A.J.)
| | - Victoria Ridger
- From the Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom (M.M.M., H.R.K., S.H., J.S.-C., S.F., N.P.B., R.M., M.A., S.E.F., T.J.A.C., V.R., P.C.E.); ERASMUS MC, Rotterdam, The Netherlands (R.X., K.v.d.H.); Vascular Biology Program, Department of Surgery (A.M.) and Department of Ophthalmology (J.C.), Boston Children's Hospital, Harvard Medical School, MA; Department of Bioengineering, Imperial College London, London, United Kingdom (P.D.W.); and Department of Cardiovascular Science, Katholieke Universiteit Leuven, Leuven, Belgium (E.A.J.)
| | - Paul C Evans
- From the Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom (M.M.M., H.R.K., S.H., J.S.-C., S.F., N.P.B., R.M., M.A., S.E.F., T.J.A.C., V.R., P.C.E.); ERASMUS MC, Rotterdam, The Netherlands (R.X., K.v.d.H.); Vascular Biology Program, Department of Surgery (A.M.) and Department of Ophthalmology (J.C.), Boston Children's Hospital, Harvard Medical School, MA; Department of Bioengineering, Imperial College London, London, United Kingdom (P.D.W.); and Department of Cardiovascular Science, Katholieke Universiteit Leuven, Leuven, Belgium (E.A.J.).
| |
Collapse
|
32
|
Wang N, Guo D, Zhao YY, Dong CY, Liu XY, Yang BX, Wang SW, Wang L, Liu QG, Ren Q, Lin YM, Ma XT. TWIST-1 promotes cell growth, drug resistance and progenitor clonogenic capacities in myeloid leukemia and is a novel poor prognostic factor in acute myeloid leukemia. Oncotarget 2016; 6:20977-92. [PMID: 26023795 PMCID: PMC4673244 DOI: 10.18632/oncotarget.4007] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 05/08/2015] [Indexed: 12/18/2022] Open
Abstract
Alterations of TWIST-1 expression are often seen in solid tumors and contribute to tumorigenesis and cancer progression. However, studies concerning its pathogenic role in leukemia are scarce. Our study shows that TWIST-1 is overexpressed in bone marrow mononuclear cells of patients with acute myeloid leukemia (AML) and chronic myeloid leukemia (CML). Gain-of-function and loss-of-function analyses demonstrate that TWIST-1 promotes cell growth, colony formation and drug resistance of AML and CML cell lines. Furthermore, TWIST-1 is aberrantly highly expressed in CD34+CD38− leukemia stem cell candidates and its expression declines with differentiation. Down-modulation of TWIST-1 in myeloid leukemia CD34+ cells impairs their colony-forming capacity. Mechanistically, c-MPL, which is highly expressed in myeloid leukemia cells and associated with poor prognosis, is identified as a TWIST-1 coexpressed gene in myeloid leukemia patients and partially contributes to TWIST-1-mediated leukemogenic effects. Moreover, patients with higher TWIST-1 expression have shorter overall and event-free survival (OS and EFS) in AML. Multivariate analysis further demonstrates that TWIST-1 overexpression is a novel independent unfavourable predictor for both OS and EFS in AML. These data highlight TWIST-1 as a new candidate gene contributing to leukemogenesis of myeloid leukemia, and propose possible new avenues for improving risk and treatment stratification in AML.
Collapse
Affiliation(s)
- Nan Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, China
| | - Dan Guo
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, China
| | - Yang-Yang Zhao
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, China
| | - Cheng-Ya Dong
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, China
| | - Xiao-Yan Liu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, China
| | - Bin-Xia Yang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, China
| | - Shu-Wei Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, China
| | - Lin Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, China
| | - Qing-Guo Liu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, China
| | - Qian Ren
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, China
| | - Yong-Min Lin
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, China
| | - Xiao-Tong Ma
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, China
| |
Collapse
|
33
|
Guo W, You X, Xu D, Zhang Y, Wang Z, Man K, Wang Z, Chen Y. PAQR3 enhances Twist1 degradation to suppress epithelial-mesenchymal transition and metastasis of gastric cancer cells. Carcinogenesis 2016; 37:397-407. [PMID: 26905590 DOI: 10.1093/carcin/bgw013] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 01/16/2016] [Indexed: 01/06/2023] Open
Abstract
Twist1 is an essential transcription factor required to initiate epithelial-mesenchymal transition (EMT) and promote tumor metastasis. PAQR3 is a newly found tumor suppressor that is frequently downregulated in many types of human cancers. Downregulation of PAQR3 is associated with accelerated metastasis and poor prognosis of the patients with gastric cancers. In this study, we demonstrate that PAQR3 is actively involved in the degradation of Twist1 and whereby regulates EMT and metastasis of gastric cancer cells. PAQR3 overexpression reduces the protein level but not the mRNA level of Twist1. The protein stability and polyubiquitination of Twist1 are altered by PAQR3. PAQR3 forms a complex with Twist1 and BTRC, an E3 ubiquitin ligase. PAQR3 enhances the interaction between Twist1 and BTRC. Twist1 is mobilized from the nucleus to a proteasome-containing structure in the cytoplasm upon overexpression of PAQR3 and BTRC, which is required for PAQR3-induced degradation of Twist1. The Twist1 box domain of the Twist1 protein is required for the interaction of Twist1 with both PAQR3 and BTRC, indispensable for PAQR3-mediated degradation of Twist1. Both BTRC and Twist1 are required for the inhibitory effects of PAQR3 on migration and EMT phenotype of gastric cancers cells. Importantly, Twist1 is indispensable for the inhibitory effect of PAQR3 on metastasis of gastric cancer cells in vivo Collectively, these findings not only pinpoint that Twist1 mediates the modulatory function of PAQR3 on EMT and metastasis but also suggest that targeting Twist1 is a promising strategy to control metastasis of tumors with downregulation of PAQR3.
Collapse
Affiliation(s)
- Weiwei Guo
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences , 320 Yueyang Road, New Life Science Building, A2214, Shanghai 200031 , China and
| | - Xue You
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, New Life Science Building, A2214, Shanghai 200031, China and.,School of Life Sciences and Technology, Shanghai Tech University, Shanghai 200031, China
| | - Daqian Xu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences , 320 Yueyang Road, New Life Science Building, A2214, Shanghai 200031 , China and
| | - Yuxue Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences , 320 Yueyang Road, New Life Science Building, A2214, Shanghai 200031 , China and
| | - Zheng Wang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences , 320 Yueyang Road, New Life Science Building, A2214, Shanghai 200031 , China and
| | - Kaiyang Man
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, New Life Science Building, A2214, Shanghai 200031, China and.,School of Life Sciences and Technology, Shanghai Tech University, Shanghai 200031, China
| | - Zhenzhen Wang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences , 320 Yueyang Road, New Life Science Building, A2214, Shanghai 200031 , China and
| | - Yan Chen
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, New Life Science Building, A2214, Shanghai 200031, China and.,School of Life Sciences and Technology, Shanghai Tech University, Shanghai 200031, China
| |
Collapse
|
34
|
Sakamoto A, Akiyama Y, Shimada S, Zhu WG, Yuasa Y, Tanaka S. DNA Methylation in the Exon 1 Region and Complex Regulation of Twist1 Expression in Gastric Cancer Cells. PLoS One 2015; 10:e0145630. [PMID: 26695186 PMCID: PMC4687923 DOI: 10.1371/journal.pone.0145630] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 12/06/2015] [Indexed: 02/06/2023] Open
Abstract
Twist1 overexpression is frequently observed in various cancers including gastric cancer (GC). Although DNA methylation of the Twist1 gene has been reported in cancer cells, the mechanisms underlying transcriptional activation remain uncertain. In this study, we first examined epigenetic alterations of the Twist1 using Twist1 transcription-positive and -negative cell lines that are derived from our established diffuse-type GC mouse model. Treatment with a DNA demethylation agent 5-aza-dC re-activated Twist1 expression in Twist1 expression-negative GC cells. According to methylation-specific PCR and bisulfite sequencing analysis, methylation at the CpG-rich region within Twist1 coding exon 1, rather than its promoter region, was tightly linked to transcriptional silencing of the Twist1 expression in mouse GC cells. Chromatin immunoprecipitation assays revealed that active histone mark H3K4me3 was enriched in Twist1 expression-positive cells, and inactive histone mark H3K9me3 was enriched in Twist1 expression-negative cells. The expression levels of Suv39h1 and Suv39h2, histone methyltransferases for H3K9me3, were inversely correlated with Twist1 expression, and knockdown of Suv39h1 or Suv39h2 induced Twist1 expression. Moreover, Sp1 transcription factor bound to the exon 1 CpG-rich region in Twist1 expression-positive cell lines, and Twist1 expression was diminished by mithramycin, which that interferes with Sp1 binding to CpG-rich regulatory sequences. Our studies suggested that the Twist1 transcription in GC cells might be regulated through potential cooperation of DNA methylation, histone modification in complex with Sp1 binding to CpG-rich regions within the exon 1 region.
Collapse
Affiliation(s)
- Ayuna Sakamoto
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshimitsu Akiyama
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- * E-mail: ;
| | - Shu Shimada
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Wei-Guo Zhu
- Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Yasuhito Yuasa
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinji Tanaka
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- * E-mail: ;
| |
Collapse
|
35
|
Quarto N, Senarath-Yapa K, Renda A, Longaker MT. TWIST1 silencing enhances in vitro and in vivo osteogenic differentiation of human adipose-derived stem cells by triggering activation of BMP-ERK/FGF signaling and TAZ upregulation. Stem Cells 2015; 33:833-47. [PMID: 25446627 DOI: 10.1002/stem.1907] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 10/06/2014] [Accepted: 10/15/2014] [Indexed: 01/10/2023]
Abstract
Mesenchymal stem cells (MSCs) show promise for cellular therapy and regenerative medicine. Human adipose tissue-derived stem cells (hASCs) represent an attractive source of seed cells in bone regeneration. How to effectively improve osteogenic differentiation of hASCs in the bone tissue engineering has become a very important question with profound translational implications. Numerous regulatory pathways dominate osteogenic differentiation of hASCs involving transcriptional factors and signaling molecules. However, how these factors combine with each other to regulate hASCs osteogenic differentiation still remains to be illustrated. The highly conserved developmental proteins TWIST play key roles for transcriptional regulation in mesenchymal cell lineages. This study investigates TWIST1 function in hASCs osteogenesis. Our results show that TWIST1 shRNA silencing increased the osteogenic potential of hASCs in vitro and their skeletal regenerative ability when applied in vivo. We demonstrate that the increased osteogenic capacity observed with TWIST1 knockdown in hASCs is mediated through endogenous activation of BMP and ERK/FGF signaling leading, in turn, to upregulation of TAZ, a transcriptional modulator of MSCs differentiation along the osteoblast lineage. Inhibition either of BMP or ERK/FGF signaling suppressed TAZ upregulation and the enhanced osteogenesis in shTWIST1 hASCs. Cosilencing of both TWIST1 and TAZ abrogated the effect elicited by TWIST1 knockdown thus, identifying TAZ as a downstream mediator through which TWIST1 knockdown enhanced osteogenic differentiation in hASCs. Our functional study contributes to a better knowledge of molecular mechanisms governing the osteogenic ability of hASCs, and highlights TWIST1 as a potential target to facilitate in vivo bone healing.
Collapse
Affiliation(s)
- Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, California, USA; Dipartimento di Scienze Biomediche Avanzate, Universita' degli Studi di Napoli Federico II, Napoli, Italy
| | | | | | | |
Collapse
|
36
|
Lin S, Ewen-Campen B, Ni X, Housden BE, Perrimon N. In Vivo Transcriptional Activation Using CRISPR/Cas9 in Drosophila. Genetics 2015; 201:433-42. [PMID: 26245833 PMCID: PMC4596659 DOI: 10.1534/genetics.115.181065] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 08/04/2015] [Indexed: 12/30/2022] Open
Abstract
A number of approaches for Cas9-mediated transcriptional activation have recently been developed, allowing target genes to be overexpressed from their endogenous genomic loci. However, these approaches have thus far been limited to cell culture, and this technique has not been demonstrated in vivo in any animal. The technique involving the fewest separate components, and therefore the most amenable to in vivo applications, is the dCas9-VPR system, where a nuclease-dead Cas9 is fused to a highly active chimeric activator domain. In this study, we characterize the dCas9-VPR system in Drosophila cells and in vivo. We show that this system can be used in cell culture to upregulate a range of target genes, singly and in multiplex, and that a single guide RNA upstream of the transcription start site can activate high levels of target transcription. We observe marked heterogeneity in guide RNA efficacy for any given gene, and we confirm that transcription is inhibited by guide RNAs binding downstream of the transcription start site. To demonstrate one application of this technique in cells, we used dCas9-VPR to identify target genes for Twist and Snail, two highly conserved transcription factors that cooperate during Drosophila mesoderm development. In addition, we simultaneously activated both Twist and Snail to identify synergistic responses to this physiologically relevant combination. Finally, we show that dCas9-VPR can activate target genes and cause dominant phenotypes in vivo, providing the first demonstration of dCas9 activation in a multicellular animal. Transcriptional activation using dCas9-VPR thus offers a simple and broadly applicable technique for a variety of overexpression studies.
Collapse
Affiliation(s)
- Shuailiang Lin
- Tsinghua-Peking-National Institute of Biological Sciences Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China Department of Genetics
| | | | | | | | - Norbert Perrimon
- Department of Genetics Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
37
|
Nuti SV, Mor G, Li P, Yin G. TWIST and ovarian cancer stem cells: implications for chemoresistance and metastasis. Oncotarget 2015; 5:7260-71. [PMID: 25238494 PMCID: PMC4202121 DOI: 10.18632/oncotarget.2428] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The transcription factor TWIST1 is a highly evolutionally conserved basic Helix-Loop-Helix (bHLH) transcription factor that functions as a master regulator of gastrulation and mesodermal development. Although TWIST1 was initially associated with embryo development, an increasing number of studies have shown TWIST1 role in the regulation of tissue homeostasis, primarily as a regulator of inflammation. More recently, TWIST1 has been found to be involved in the process of tumor metastasis through the regulation of Epithelial Mesenchymal Transition (EMT). The objective of this review is to examine the normal functions of TWIST1 and its role in tumor development, with a particular focus on ovarian cancer. We discuss the potential role of TWIST1 in the context of ovarian cancer stem cells and its influence in the process of tumor formation.
Collapse
Affiliation(s)
- Sudhakar V Nuti
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Gil Mor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Peiyao Li
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Gang Yin
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
38
|
Sachs L, Chen YT, Drechsler A, Lynch JA, Panfilio KA, Lässig M, Berg J, Roth S. Dynamic BMP signaling polarized by Toll patterns the dorsoventral axis in a hemimetabolous insect. eLife 2015; 4:e05502. [PMID: 25962855 PMCID: PMC4423117 DOI: 10.7554/elife.05502] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 04/12/2015] [Indexed: 11/13/2022] Open
Abstract
Toll-dependent patterning of the dorsoventral axis in Drosophila represents one of the best understood gene regulatory networks. However, its evolutionary origin has remained elusive. Outside the insects Toll is not known for a patterning function, but rather for a role in pathogen defense. Here, we show that in the milkweed bug Oncopeltus fasciatus, whose lineage split from Drosophila's more than 350 million years ago, Toll is only required to polarize a dynamic BMP signaling network. A theoretical model reveals that this network has self-regulatory properties and that shallow Toll signaling gradients are sufficient to initiate axis formation. Such gradients can account for the experimentally observed twinning of insect embryos upon egg fragmentation and might have evolved from a state of uniform Toll activity associated with protecting insect eggs against pathogens.
Collapse
Affiliation(s)
- Lena Sachs
- Institute for Developmental Biology, University of Cologne, Köln, Germany
| | - Yen-Ta Chen
- Institute for Developmental Biology, University of Cologne, Köln, Germany
| | - Axel Drechsler
- Institute for Developmental Biology, University of Cologne, Köln, Germany
- Bundesministerium für Umwelt, Naturschutz, Bau und Reaktorsicherheit, Bonn, Germany
| | - Jeremy A Lynch
- Institute for Developmental Biology, University of Cologne, Köln, Germany
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, United States
| | - Kristen A Panfilio
- Institute for Developmental Biology, University of Cologne, Köln, Germany
| | - Michael Lässig
- Institute for Theoretical Physics, University of Cologne, Cologne, Germany
| | - Johannes Berg
- Institute for Theoretical Physics, University of Cologne, Cologne, Germany
| | - Siegfried Roth
- Institute for Developmental Biology, University of Cologne, Köln, Germany
| |
Collapse
|
39
|
Polyakov O, He B, Swan M, Shaevitz JW, Kaschube M, Wieschaus E. Passive mechanical forces control cell-shape change during Drosophila ventral furrow formation. Biophys J 2015; 107:998-1010. [PMID: 25140436 DOI: 10.1016/j.bpj.2014.07.013] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 07/02/2014] [Accepted: 07/07/2014] [Indexed: 11/16/2022] Open
Abstract
During Drosophila gastrulation, the ventral mesodermal cells constrict their apices, undergo a series of coordinated cell-shape changes to form a ventral furrow (VF) and are subsequently internalized. Although it has been well documented that apical constriction is necessary for VF formation, the mechanism by which apical constriction transmits forces throughout the bulk tissue of the cell remains poorly understood. In this work, we develop a computational vertex model to investigate the role of the passive mechanical properties of the cellular blastoderm during gastrulation. We introduce to our knowledge novel data that confirm that the volume of apically constricting cells is conserved throughout the entire course of invagination. We show that maintenance of this constant volume is sufficient to generate invagination as a passive response to apical constriction when it is combined with region-specific elasticities in the membranes surrounding individual cells. We find that the specific sequence of cell-shape changes during VF formation is critically controlled by the stiffness of the lateral and basal membrane surfaces. In particular, our model demonstrates that a transition in basal rigidity is sufficient to drive VF formation along the same sequence of cell-shape change that we observed in the actual embryo, with no active force generation required other than apical constriction.
Collapse
Affiliation(s)
- Oleg Polyakov
- Department of Physics, Princeton University, Princeton University, Princeton, New Jersey.
| | - Bing He
- Department of Molecular Biology, Princeton University, Princeton, New Jersey
| | - Michael Swan
- Department of Molecular Biology, Princeton University, Princeton, New Jersey
| | - Joshua W Shaevitz
- Department of Physics, Princeton University, Princeton University, Princeton, New Jersey
| | - Matthias Kaschube
- Frankfurt Institute for Advanced Studies, Faculty of Computer Science and Mathematics, Goethe University, Frankfurt am Main, Germany
| | - Eric Wieschaus
- Department of Molecular Biology, Princeton University, Princeton, New Jersey; Howard Hughes Medical Institute, Princeton University, Princeton, New Jersey
| |
Collapse
|
40
|
Lin HS, Gong JN, Su R, Chen MT, Song L, Shen C, Wang F, Ma YN, Zhao HL, Yu J, Li WW, Huang LX, Xu XH, Zhang JW. miR-199a-5p inhibits monocyte/macrophage differentiation by targeting the activin A type 1B receptor gene and finally reducing C/EBPα expression. J Leukoc Biol 2014; 96:1023-35. [PMID: 25258381 DOI: 10.1189/jlb.1a0514-240r] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
miRNAs are short, noncoding RNAs that regulate expression of target genes at post-transcriptional levels and function in many important cellular processes, including differentiation, proliferation, etc. In this study, we observed down-regulation of miR-199a-5p during monocyte/macrophage differentiation of HL-60 and THP-1 cells, as well as human CD34(+) HSPCs. This down-regulation of miR-199a-5p resulted from the up-regulation of PU.1 that was demonstrated to regulate transcription of the miR-199a-2 gene negatively. Overexpression of miR-199a-5p by miR-199a-5p mimic transfection or lentivirus-mediated gene transfer significantly inhibited monocyte/macrophage differentiation of the cell lines or HSPCs. The mRNA encoding an ACVR1B was identified as a direct target of miR-199a-5p. Gradually increased ACVR1B expression level was detected during monocyte/macrophage differentiation of the leukemic cell lines and HSPCs, and knockdown of ACVR1B resulted in inhibition of monocyte/macrophage differentiation of HL-60 and THP-1 cells, which suggested that ACVR1B functions as a positive regulator of monocyte/macrophage differentiation. We demonstrated that miR-199a-5p overexpression or ACVR1B knockdown promoted proliferation of THP-1 cells through increasing phosphorylation of Rb. We also demonstrated that the down-regulation of ACVR1B reduced p-Smad2/3, which resulted in decreased expression of C/EBPα, a key regulator of monocyte/macrophage differentiation, and finally, inhibited monocyte/macrophage differentiation.
Collapse
Affiliation(s)
- Hai-Shuang Lin
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia-Nan Gong
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rui Su
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming-Tai Chen
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Song
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Shen
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fang Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan-Ni Ma
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hua-Lu Zhao
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia Yu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei-Wei Li
- Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Li-Xia Huang
- First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China; and
| | - Xin-Hua Xu
- Taizhou Cancer Hospital, Zhejiang Province, China
| | - Jun-Wu Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China;
| |
Collapse
|
41
|
Caine C, Kasherov P, Silber J, Lalouette A. Mef2 interacts with the Notch pathway during adult muscle development in Drosophila melanogaster. PLoS One 2014; 9:e108149. [PMID: 25247309 PMCID: PMC4172597 DOI: 10.1371/journal.pone.0108149] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 08/03/2014] [Indexed: 12/22/2022] Open
Abstract
Myogenesis of indirect flight muscles (IFMs) in Drosophila melanogaster follows a well-defined cellular developmental scheme. During embryogenesis, a set of cells, the Adult Muscle Precursors (AMPs), are specified. These cells will become proliferating myoblasts during the larval stages which will then give rise to the adult IFMs. Although the cellular aspect of this developmental process is well studied, the molecular biology behind the different stages is still under investigation. In particular, the interactions required during the transition from proliferating myoblasts to differentiated myoblasts ready to fuse to the muscle fiber. It has been previously shown that the Notch pathway is active in proliferating myoblasts, and that this pathway is inhibited in developing muscle fibers. Furthermore, the Myocyte Enhancing Factor 2 (Mef2), Vestigial (Vg) and Scalloped (Sd) transcription factors are necessary for IFM development and that Vg is required for Notch pathway repression in differentiating fibers. Here we examine the interactions between Notch and Mef2 and mechanisms by which the Notch pathway is inhibited during differentiation. We show that Mef2 is capable of inhibiting the Notch pathway in non myogenic cells. A previous screen for Mef2 potential targets identified Delta a component of the Notch pathway. Dl is expressed in Mef2 and Sd-positive developing fibers. Our results show that Mef2 and possibly Sd regulate a Dl enhancer specifically expressed in the developing IFMs and that Mef2 is required for Dl expression in developing IFMs.
Collapse
Affiliation(s)
- Charlotte Caine
- Institut Jacques Monod, CNRS, UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Petar Kasherov
- Institut Jacques Monod, CNRS, UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Joël Silber
- Institut Jacques Monod, CNRS, UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Alexis Lalouette
- Institut Jacques Monod, CNRS, UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- * E-mail:
| |
Collapse
|
42
|
Gunage RD, Reichert H, VijayRaghavan K. Identification of a new stem cell population that generates Drosophila flight muscles. eLife 2014; 3:e03126. [PMID: 25135939 PMCID: PMC4171707 DOI: 10.7554/elife.03126] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 08/15/2014] [Indexed: 12/20/2022] Open
Abstract
How myoblast populations are regulated for the formation of muscles of different sizes is an essentially unanswered question. The large flight muscles of Drosophila develop from adult muscle progenitor (AMP) cells set-aside embryonically. The thoracic segments are all allotted the same small AMP number, while those associated with the wing-disc proliferate extensively to give rise to over 2500 myoblasts. An initial amplification occurs through symmetric divisions and is followed by a switch to asymmetric divisions in which the AMPs self-renew and generate post-mitotic myoblasts. Notch signaling controls the initial amplification of AMPs, while the switch to asymmetric division additionally requires Wingless, which regulates Numb expression in the AMP lineage. In both cases, the epidermal tissue of the wing imaginal disc acts as a niche expressing the ligands Serrate and Wingless. The disc-associated AMPs are a novel muscle stem cell population that orchestrates the early phases of adult flight muscle development.
Collapse
Affiliation(s)
- Rajesh D Gunage
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | | | - K VijayRaghavan
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| |
Collapse
|
43
|
Zhang X, Ma W, Cui J, Yao H, Zhou H, Ge Y, Xiao L, Hu X, Liu BH, Yang J, Li YY, Chen S, Eaves CJ, Wu D, Zhao Y. Regulation of p21 by TWIST2 contributes to its tumor-suppressor function in human acute myeloid leukemia. Oncogene 2014; 34:3000-10. [DOI: 10.1038/onc.2014.241] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 06/08/2014] [Accepted: 06/15/2014] [Indexed: 12/18/2022]
|
44
|
Tang YL, Jiang J, Liu J, Zheng M, He YW, Chen W, Fan YL, Chen QM, Liao CH, Liang XH. Hyperthermia inhibited the migration of tongue squamous cell carcinoma through TWIST2. J Oral Pathol Med 2014; 44:337-44. [PMID: 25048179 DOI: 10.1111/jop.12237] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2014] [Indexed: 02/05/2023]
Abstract
BACKGROUND Hyperthermia has been shown promising in the treatment of head and neck squamous cell carcinoma (HNSCC); however, the mechanism underlying hyperthermia reducing tumor metastasis is poorly elucidated. TWIST2, an important transcription factor of epithelial-mesenchymal transition (EMT), plays a critical role in the tumor progression and metastasis. The role of TWIST2 in tongue squamous cell carcinoma (TSCC) and its association with hyperthermia still have not been reported. METHOD The correlations between TWIST2 expression and the clinical-pathologic characteristics of 89 patients with TSCC were evaluated by immunohistochemical staining. TSCC cell lines transfected with siRNA against TWIST2 were heated for 40 min at 42.5°C, and the migration capability of cells was examined by migration assay. Xenograft tumors in nude mice were treated by hyperthermia, and TWIST2 expression was measured. RESULTS Our data showed that TWIST2 expression was associated with the metastasis of human TSCC. In Tca8113 and Cal-27 cells, TWIST2-siRNA treatment can reduce cell migration ability and has no effect on the cell proliferation and apoptosis. Hyperthermia can decrease the level of TWIST2 in TSCC and inhibit the migration of cells. CONCLUSIONS This demonstrated that hyperthermia might decrease the migration of Tca8113 and Cal-27 cells by reducing TWIST2 expression. Altogether, these findings suggest an as yet undescribed link between TWIST2 and hyperthermia in TSCC.
Collapse
Affiliation(s)
- Ya-ling Tang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, (Sichuan University), Chengdu, Sichuan, China; Department of Oral Pathology, West China Hospital of Stomatology, (Sichuan University), Chengdu, Sichuan, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Rembold M, Ciglar L, Yáñez-Cuna JO, Zinzen RP, Girardot C, Jain A, Welte MA, Stark A, Leptin M, Furlong EEM. A conserved role for Snail as a potentiator of active transcription. Genes Dev 2014; 28:167-81. [PMID: 24402316 PMCID: PMC3909790 DOI: 10.1101/gad.230953.113] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The transcription factors of the Snail family are key regulators of epithelial-mesenchymal transitions, cell morphogenesis, and tumor metastasis. Since its discovery in Drosophila ∼25 years ago, Snail has been extensively studied for its role as a transcriptional repressor. Here we demonstrate that Drosophila Snail can positively modulate transcriptional activation. By combining information on in vivo occupancy with expression profiling of hand-selected, staged snail mutant embryos, we identified 106 genes that are potentially directly regulated by Snail during mesoderm development. In addition to the expected Snail-repressed genes, almost 50% of Snail targets showed an unanticipated activation. The majority of "Snail-activated" genes have enhancer elements cobound by Twist and are expressed in the mesoderm at the stages of Snail occupancy. Snail can potentiate Twist-mediated enhancer activation in vitro and is essential for enhancer activity in vivo. Using a machine learning approach, we show that differentially enriched motifs are sufficient to predict Snail's regulatory response. In silico mutagenesis revealed a likely causative motif, which we demonstrate is essential for enhancer activation. Taken together, these data indicate that Snail can potentiate enhancer activation by collaborating with different activators, providing a new mechanism by which Snail regulates development.
Collapse
Affiliation(s)
- Martina Rembold
- Institute of Genetics, University of Cologne, 50674 Cologne, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Ratnaparkhi A. Signaling by Folded gastrulation is modulated by mitochondrial fusion and fission. J Cell Sci 2013; 126:5369-76. [PMID: 24101729 DOI: 10.1242/jcs.127985] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mitochondria are increasingly being identified as integrators and regulators of cell signaling pathways. Folded gastrulation (Fog) is a secreted signaling molecule best known for its role in regulating cell shape change at the ventral furrow during gastrulation in Drosophila. Fog is thought to signal, through a G-protein-coupled receptor, to effect downstream cytoskeletal changes necessary for cell shape change. However, the mechanisms regulating Fog signaling that lead to change in cell morphology are poorly understood. This study describes the identification of proteins involved in mitochondrial fusion and fission as regulators of Fog signaling. Pro-fission factors were found to function as enhancers of signaling, whereas pro-fusion factors were found to have the opposite effect. Consistent with this, activation of Fog signaling resulted in mitochondrial fragmentation, and inhibiting this process could attenuate Fog signaling. The findings presented here show that mitochondria, through regulation of fusion and fission, function as downstream effectors and modulators of Fog signaling and Fog-dependent cell shape change.
Collapse
Affiliation(s)
- Anuradha Ratnaparkhi
- Agharkar Research Institute, Animal Sciences Division, Zoology Group, G.G. Agarkar Road, Pune 411 004, India
| |
Collapse
|
47
|
Li LZ, Zhang CZ, Liu LL, Yi C, Lu SX, Zhou X, Zhang ZJ, Peng YH, Yang YZ, Yun JP. miR-720 inhibits tumor invasion and migration in breast cancer by targeting TWIST1. Carcinogenesis 2013; 35:469-78. [DOI: 10.1093/carcin/bgt330] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
48
|
Pfeifer K, Schaub C, Wolfstetter G, Dorresteijn A. Identification and characterization of a twist ortholog in the polychaete annelid Platynereis dumerilii reveals mesodermal expression of Pdu-twist. Dev Genes Evol 2013; 223:319-28. [PMID: 23817621 DOI: 10.1007/s00427-013-0448-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 06/11/2013] [Indexed: 01/26/2023]
Abstract
The basic helix-loop-helix transcription factor twist plays a key role during mesoderm development in Bilateria. In this study, we identified a twist ortholog in the polychaete annelid Platynereis dumerilii and analyze its expression during larval development, postlarval growth up to the adult stage, and caudal regeneration after amputation of posterior segments. At late larval stages, Pdu-twist is expressed in the mesodermal anlagen and in developing muscles. During adulthood and caudal regeneration, Pdu-twist is expressed in the posterior growth zone, in mesodermal cells within the newly forming segments and budding parapodia. Our results indicate that Pdu-twist is involved in mesoderm formation during larval development, posterior growth, and caudal regeneration.
Collapse
Affiliation(s)
- Kathrin Pfeifer
- Institut für Allgemeine und Spezielle Zoologie, Allgemeine Zoologie und Entwicklungsbiologie, Justus-Liebig-Universität Giessen, Germany
| | | | | | | |
Collapse
|
49
|
Ren KQ, Cao XZ, Liu ZH, Guo H, Quan MF, Liu F, Jiang L, Xiang HL, Deng XY, Cao JG. 8-bromo-5-hydroxy-7-methoxychrysin targeting for inhibition of the properties of liver cancer stem cells by modulation of Twist signaling. Int J Oncol 2013; 43:1719-29. [PMID: 23970349 DOI: 10.3892/ijo.2013.2071] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 07/28/2013] [Indexed: 11/06/2022] Open
Abstract
Emerging evidence has suggested that cancer stem cells with expression of surface biomarkers including CD133 and CD44 have more aggressive biological behavior, including epithelial-mesenchymal transition (EMT), which are closely related to invasion. The upregulation and nuclear relocation of the EMT regulator Twist1 have been implicated in the tumor invasion and metastasis of human hepatocellular carcinoma (HCC). In this study, we aimed to isolate and characterize a small population of CD133+ cells that existed in the HCC cell line SMMC-7721 by MACS and investigated the possible roles of 8-bromo-7-methoxychrysin (BrMC), a synthetic analogue of chrysin, in inhibiting the properties of CD133+ sphere-forming cells (SFCs) derived from the HCC cell line SMMC-7721, namely liver cancer stem cells (LCSCs). Based on the data, BrMC inhibited the proliferation, self-renewal and invasion of LCSCs in vitro and in vivo, downregulated the expression of the LCSC biomarkers CD133 and CD44 and induced EMT by downregulating the expression of Twist and β-catenin in LCSCs. BrMC potentiated the inhibition of LCSCs self-renewal after reduction of twist protein levels, which was attenuated when twist was overexpressed. This study not only provides an important experimental and theoretical basis for investigation of BrMC in LCSCs, but also helps in the development of effective therapeutic medicine for HCC.
Collapse
Affiliation(s)
- Kai-Qun Ren
- Medical College, Hunan Normal University, Changsha, Hunan 410013, P.R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Microarray comparison of anterior and posterior Drosophila wing imaginal disc cells identifies novel wing genes. G3-GENES GENOMES GENETICS 2013; 3:1353-62. [PMID: 23749451 PMCID: PMC3737175 DOI: 10.1534/g3.113.006569] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Signaling between cells in the anterior (A) and posterior (P) compartments directs Drosophila wing disc development and is dependent on expression of the homeodomain transcription factor Engrailed (En) in P cells. Downstream of en, posteriorly expressed Hedgehog (Hh) protein signals across the A/P border to establish a developmental organizer that directs pattern formation and growth throughout the wing primordium. Here we extend investigations of the processes downstream of en by using expression array analysis to compare A and P cells. A total of 102 candidate genes were identified that express differentially in the A and P compartments; four were characterized: Stubble (Sb) expression is restricted to A cells due to repression by en. CG15905, CG16884; CG10200/hase und igel (hui) are expressed in A cells downstream of Hh signaling; and RNA interference for hui, Stubble, and CG16884 revealed that each is essential to wing development.
Collapse
|