1
|
Cao Y. Lack of basic rationale in epithelial-mesenchymal transition and its related concepts. Cell Biosci 2024; 14:104. [PMID: 39164745 PMCID: PMC11334496 DOI: 10.1186/s13578-024-01282-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/05/2024] [Indexed: 08/22/2024] Open
Abstract
Epithelial-mesenchymal transition (EMT) is defined as a cellular process during which epithelial cells acquire mesenchymal phenotypes and behavior following the downregulation of epithelial features. EMT and its reversed process, the mesenchymal-epithelial transition (MET), and the special form of EMT, the endothelial-mesenchymal transition (EndMT), have been considered as mainstream concepts and general rules driving developmental and pathological processes, particularly cancer. However, discrepancies and disputes over EMT and EMT research have also grown over time. EMT is defined as transition between two cellular states, but it is unanimously agreed by EMT researchers that (1) neither the epithelial and mesenchymal states nor their regulatory networks have been clearly defined, (2) no EMT markers or factors can represent universally epithelial and mesenchymal states, and thus (3) EMT cannot be assessed on the basis of one or a few EMT markers. In contrast to definition and proposed roles of EMT, loss of epithelial feature does not cause mesenchymal phenotype, and EMT does not contribute to embryonic mesenchyme and neural crest formation, the key developmental events from which the EMT concept was derived. EMT and MET, represented by change in cell shapes or adhesiveness, or symbolized by EMT factors, are biased interpretation of the overall change in cellular property and regulatory networks during development and cancer progression. Moreover, EMT and MET are consequences rather than driving factors of developmental and pathological processes. The true meaning of EMT in some developmental and pathological processes, such as fibrosis, needs re-evaluation. EMT is believed to endow malignant features, such as migration, stemness, etc., to cancer cells. However, the core property of cancer (tumorigenic) cells is neural stemness, and the core EMT factors are components of the regulatory networks of neural stemness. Thus, EMT in cancer progression is misattribution of the roles of neural stemness to the unknown mesenchymal state. Similarly, neural crest EMT is misattribution of intrinsic property of neural crest cells to the unknown mesenchymal state. Lack of basic rationale in EMT and related concepts urges re-evaluation of their significance as general rules for understanding developmental and pathological processes, and re-evaluation of their significance in scientific research.
Collapse
Affiliation(s)
- Ying Cao
- The MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Medical School of Nanjing University, 12 Xuefu Road, Pukou High-Tech Zone, Nanjing, 210061, China.
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China.
- Shenzhen Research Institute of Nanjing University, Shenzhen, China.
| |
Collapse
|
2
|
Seto Y, Ogihara R, Takizawa K, Eiraku M. In vitro induction of patterned branchial arch-like aggregate from human pluripotent stem cells. Nat Commun 2024; 15:1351. [PMID: 38355589 PMCID: PMC10867012 DOI: 10.1038/s41467-024-45285-0] [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: 05/16/2023] [Accepted: 01/19/2024] [Indexed: 02/16/2024] Open
Abstract
Early patterning of neural crest cells (NCCs) in the craniofacial primordium is important for subsequent development of proper craniofacial structures. However, because of the complexity of the environment of developing tissues, surveying the early specification and patterning of NCCs is difficult. In this study, we develop a simplified in vitro 3D model using human pluripotent stem cells to analyze the early stages of facial development. In this model, cranial NCC-like cells spontaneously differentiate from neural plate border-like cells into maxillary arch-like mesenchyme after a long-term culture. Upon the addition of EDN1 and BMP4, these aggregates are converted into a mandibular arch-like state. Furthermore, temporary treatment with EDN1 and BMP4 induces the formation of spatially separated domains expressing mandibular and maxillary arch markers within a single aggregate. These results suggest that this in vitro model is useful for determining the mechanisms underlying cell fate specification and patterning during early facial development.
Collapse
Affiliation(s)
- Yusuke Seto
- Laboratory of Developmental Systems, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan.
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan.
| | - Ryoma Ogihara
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan
| | - Kaori Takizawa
- Laboratory of Developmental Systems, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Mototsugu Eiraku
- Laboratory of Developmental Systems, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan.
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan.
- Institute for Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan.
| |
Collapse
|
3
|
Zhang YY, Li F, Zeng XK, Zou YH, Zhu BB, Ye JJ, Zhang YX, Jin Q, Nie X. Single cell RNA sequencing reveals mesenchymal heterogeneity and critical functions of Cd271 in tooth development. World J Stem Cells 2023; 15:589-606. [PMID: 37424952 PMCID: PMC10324503 DOI: 10.4252/wjsc.v15.i6.589] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/18/2023] [Accepted: 05/05/2023] [Indexed: 06/20/2023] Open
Abstract
BACKGROUND Accumulating evidence suggests that the maxillary process, to which cranial crest cells migrate, is essential to tooth development. Emerging studies indicate that Cd271 plays an essential role in odontogenesis. However, the underlying mechanisms have yet to be elucidated.
AIM To establish the functionally heterogeneous population in the maxillary process, elucidate the effects of Cd271 deficiency on gene expression differences.
METHODS p75NTR knockout (Cd271-/-) mice (from American Jackson laboratory) were used to collect the maxillofacial process tissue of p75NTR knockout mice, and the wild-type maxillofacial process of the same pregnant mouse wild was used as control. After single cell suspension, the cDNA was prepared by loading the single cell suspension into the 10x Genomics Chromium system to be sequenced by NovaSeq6000 sequencing system. Finally, the sequencing data in Fastq format were obtained. The FastQC software is used to evaluate the quality of data and CellRanger analyzed the data. The gene expression matrix is read by R software, and Seurat is used to control and standardize the data, reduce the dimension and cluster. We search for marker genes for subgroup annotation by consulting literature and database; explore the effect of p75NTR knockout on mesenchymal stem cells (MSCs) gene expression and cell proportion by cell subgrouping, differential gene analysis, enrichment analysis and protein-protein interaction network analysis; understand the interaction between MSCs cells and the differentiation trajectory and gene change characteristics of p75NTR knockout MSCs by cell communication analysis and pseudo-time analysis. Last we verified the findings single cell sequencing in vitro.
RESULTS We identified 21 cell clusters, and we re-clustered these into three subclusters. Importantly, we revealed the cell–cell communication networks between clusters. We clarified that Cd271 was significantly associated with the regulation of mineralization.
CONCLUSION This study provides comprehensive mechanistic insights into the maxillary- process-derived MSCs and demonstrates that Cd271 is significantly associated with the odontogenesis in mesenchymal populations.
Collapse
Affiliation(s)
- Yan-Yan Zhang
- School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Feng Li
- School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Xiao-Ke Zeng
- School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Yan-Hui Zou
- School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Bing-Bing Zhu
- School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Jia-Jia Ye
- School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Yun-Xiao Zhang
- School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Qiu Jin
- School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Xin Nie
- School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| |
Collapse
|
4
|
Zhao R, Trainor PA. Epithelial to mesenchymal transition during mammalian neural crest cell delamination. Semin Cell Dev Biol 2022; 138:54-67. [PMID: 35277330 DOI: 10.1016/j.semcdb.2022.02.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 02/08/2022] [Accepted: 02/21/2022] [Indexed: 11/18/2022]
Abstract
Epithelial to mesenchymal transition (EMT) is a well-defined cellular process that was discovered in chicken embryos and described as "epithelial to mesenchymal transformation" [1]. During EMT, epithelial cells lose their epithelial features and acquire mesenchymal character with migratory potential. EMT has subsequently been shown to be essential for both developmental and pathological processes including embryo morphogenesis, wound healing, tissue fibrosis and cancer [2]. During the past 5 years, interest and study of EMT especially in cancer biology have increased exponentially due to the implied role of EMT in multiple aspects of malignancy such as cell invasion, survival, stemness, metastasis, therapeutic resistance and tumor heterogeneity [3]. Since the process of EMT in embryogenesis and cancer progression shares similar phenotypic changes, core transcription factors and molecular mechanisms, it has been proposed that the initiation and development of carcinoma could be attributed to abnormal activation of EMT factors usually required for normal embryo development. Therefore, developmental EMT mechanisms, whose timing, location, and tissue origin are strictly regulated, could prove useful for uncovering new insights into the phenotypic changes and corresponding gene regulatory control of EMT under pathological conditions. In this review, we initially provide an overview of the phenotypic and molecular mechanisms involved in EMT and discuss the newly emerging concept of epithelial to mesenchymal plasticity (EMP). Then we focus on our current knowledge of a classic developmental EMT event, neural crest cell (NCC) delamination, highlighting key differences in our understanding of NCC EMT between mammalian and non-mammalian species. Lastly, we highlight available tools and future directions to advance our understanding of mammalian NCC EMT.
Collapse
Affiliation(s)
- Ruonan Zhao
- Stowers Institute for Medical Research, Kansas City, MO, USA; Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, MO, USA; Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
5
|
Brenner AK, Gunnes MW. Therapeutic Targeting of the Anaplastic Lymphoma Kinase (ALK) in Neuroblastoma-A Comprehensive Update. Pharmaceutics 2021; 13:pharmaceutics13091427. [PMID: 34575503 PMCID: PMC8470592 DOI: 10.3390/pharmaceutics13091427] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/27/2021] [Accepted: 08/29/2021] [Indexed: 01/27/2023] Open
Abstract
Neuroblastoma (NBL) is an embryonic malignancy of the sympathetic nervous system and mostly affects children under the age of five. NBL is highly heterogeneous and ranges from spontaneously regressing to highly aggressive disease. One of the risk factors for poor prognosis are aberrations in the receptor tyrosine kinase anaplastic lymphoma kinase (ALK), which is involved in the normal development and function of the nervous system. ALK mutations lead to constitutive activation of ALK and its downstream signalling pathways, thus driving tumorigenesis. A wide range of steric ALK inhibitors has been synthesized, and several of these inhibitors are already in clinical use. Major challenges are acquired drug resistance to steric inhibitors and pathway evasion strategies of cancer cells upon targeted therapy. This review will give a comprehensive overview on ALK inhibitors in clinical use in high-risk NBL and on the potential and limitations of novel inhibitors. Because combinatory treatment regimens are probably less likely to induce drug resistance, a special focus will be on the combination of ALK inhibitors with drugs that either target downstream signalling pathways or that affect the survival and proliferation of cancer cells in general.
Collapse
|
6
|
PKCα Inhibition as a Strategy to Sensitize Neuroblastoma Stem Cells to Etoposide by Stimulating Ferroptosis. Antioxidants (Basel) 2021; 10:antiox10050691. [PMID: 33924765 PMCID: PMC8145544 DOI: 10.3390/antiox10050691] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/19/2021] [Accepted: 04/24/2021] [Indexed: 12/18/2022] Open
Abstract
Cancer stem cells (CSCs) are a limited cell population inside a tumor bulk characterized by high levels of glutathione (GSH), the most important antioxidant thiol of which cysteine is the limiting amino acid for GSH biosynthesis. In fact, CSCs over-express xCT, a cystine transporter stabilized on cell membrane through interaction with CD44, a stemness marker whose expression is modulated by protein kinase Cα (PKCα). Since many chemotherapeutic drugs, such as Etoposide, exert their cytotoxic action by increasing reactive oxygen species (ROS) production, the presence of high antioxidant defenses confers to CSCs a crucial role in chemoresistance. In this study, Etoposide-sensitive and -resistant neuroblastoma CSCs were chronically treated with Etoposide, given alone or in combination with Sulfasalazine (SSZ) or with an inhibitor of PKCα (C2-4), which target xCT directly or indirectly, respectively. Both combined approaches are able to sensitize CSCs to Etoposide by decreasing intracellular GSH levels, inducing a metabolic switch from OXPHOS to aerobic glycolysis, down-regulating glutathione-peroxidase-4 activity and stimulating lipid peroxidation, thus leading to ferroptosis. Our results suggest, for the first time, that PKCα inhibition inducing ferroptosis might be a useful strategy with which to fight CSC chemoresistance.
Collapse
|
7
|
Kobayashi GS, Musso CM, Moreira DDP, Pontillo-Guimarães G, Hsia GSP, Caires-Júnior LC, Goulart E, Passos-Bueno MR. Recapitulation of Neural Crest Specification and EMT via Induction from Neural Plate Border-like Cells. Stem Cell Reports 2020; 15:776-788. [PMID: 32857981 PMCID: PMC7486307 DOI: 10.1016/j.stemcr.2020.07.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022] Open
Abstract
Neural crest cells (NCCs) contribute to several tissues during embryonic development. NCC formation depends on activation of tightly regulated molecular programs at the neural plate border (NPB) region, which initiate NCC specification and epithelial-to-mesenchymal transition (EMT). Although several approaches to investigate NCCs have been devised, these early events of NCC formation remain largely unknown in humans, and currently available cellular models have not investigated EMT. Here, we report that the E6 neural induction protocol converts human induced pluripotent stem cells into NPB-like cells (NBCs), from which NCCs can be efficiently derived. NBC-to-NCC induction recapitulates gene expression dynamics associated with NCC specification and EMT, including downregulation of NPB factors and upregulation of NCC specifiers, coupled with other EMT-associated cell-state changes, such as cadherin modulation and activation of TWIST1 and other EMT inducers. This strategy will be useful in future basic or translational research focusing on these early steps of NCC formation.
Collapse
Affiliation(s)
- Gerson Shigeru Kobayashi
- Centro de Pesquisa sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil.
| | - Camila Manso Musso
- Centro de Pesquisa sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Danielle de Paula Moreira
- Centro de Pesquisa sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Giovanna Pontillo-Guimarães
- Centro de Pesquisa sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Gabriella Shih Ping Hsia
- Centro de Pesquisa sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Luiz Carlos Caires-Júnior
- Centro de Pesquisa sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Ernesto Goulart
- Centro de Pesquisa sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Maria Rita Passos-Bueno
- Centro de Pesquisa sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
8
|
Dunkel H, Chaverra M, Bradley R, Lefcort F. FGF
signaling is required for chemokinesis and ventral migration of trunk neural crest cells. Dev Dyn 2020; 249:1077-1097. [DOI: 10.1002/dvdy.190] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 04/24/2020] [Accepted: 05/04/2020] [Indexed: 11/08/2022] Open
Affiliation(s)
- Haley Dunkel
- Department of Cell Biology and NeuroscienceMontana State University Bozeman Montana USA
| | - Martha Chaverra
- Department of Cell Biology and NeuroscienceMontana State University Bozeman Montana USA
| | - Roger Bradley
- Department of Cell Biology and NeuroscienceMontana State University Bozeman Montana USA
| | - Frances Lefcort
- Department of Cell Biology and NeuroscienceMontana State University Bozeman Montana USA
| |
Collapse
|
9
|
Nasoori A. Formation, structure, and function of extra-skeletal bones in mammals. Biol Rev Camb Philos Soc 2020; 95:986-1019. [PMID: 32338826 DOI: 10.1111/brv.12597] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 03/07/2020] [Accepted: 03/17/2020] [Indexed: 12/12/2022]
Abstract
This review describes the formation, structure, and function of bony compartments in antlers, horns, ossicones, osteoderm and the os penis/os clitoris (collectively referred to herein as AHOOO structures) in extant mammals. AHOOOs are extra-skeletal bones that originate from subcutaneous (dermal) tissues in a wide variety of mammals, and this review elaborates on the co-development of the bone and skin in these structures. During foetal stages, primordial cells for the bony compartments arise in subcutaneous tissues. The epithelial-mesenchymal transition is assumed to play a key role in the differentiation of bone, cartilage, skin and other tissues in AHOOO structures. AHOOO ossification takes place after skeletal bone formation, and may depend on sexual maturity. Skin keratinization occurs in tandem with ossification and may be under the control of androgens. Both endochondral and intramembranous ossification participate in bony compartment formation. There is variation in gradients of density in different AHOOO structures. These gradients, which vary according to function and species, primarily reduce mechanical stress. Anchorage of AHOOOs to their surrounding tissues fortifies these structures and is accomplished by bone-bone fusion and Sharpey fibres. The presence of the integument is essential for the protection and function of the bony compartments. Three major functions can be attributed to AHOOOs: mechanical, visual, and thermoregulatory. This review provides the first extensive comparative description of the skeletal and integumentary systems of AHOOOs in a variety of mammals.
Collapse
Affiliation(s)
- Alireza Nasoori
- School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Hokkaido, 060-0818, Japan
| |
Collapse
|
10
|
Lee VM, Hernandez S, Giang B, Chabot C, Hernandez J, de Bellard ME. Molecular Events Controlling Cessation of Trunk Neural Crest Migration and Onset of Differentiation. Front Cell Dev Biol 2020; 8:199. [PMID: 32318567 PMCID: PMC7147452 DOI: 10.3389/fcell.2020.00199] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 03/09/2020] [Indexed: 12/30/2022] Open
Abstract
Neural crest cells (NCC) migrate extensively in vertebrate embryos to populate diverse derivatives including ganglia of the peripheral nervous system. Little is known about the molecular mechanisms that lead migrating trunk NCC to settle at selected sites in the embryo, ceasing their migration and initiating differentiation programs. To identify candidate genes involved in these processes, we profiled genes up-regulated in purified post-migratory compared with migratory NCC using a staged, macroarrayed cDNA library. A secondary screen of in situ hybridization revealed that many genes are specifically enhanced in neural crest-derived ganglia, including macrophage migration inhibitory factor (MIF), a ligand for CXCR4 receptor. Through in vivo and in vitro assays, we found that MIF functions as a potent chemoattractant for NCC. These results provide a molecular profile of genes expressed concomitant with gangliogenesis, thus, offering new markers and potential regulatory candidates involved in cessation of migration and onset of differentiation.
Collapse
Affiliation(s)
- Vivian M Lee
- Universal Cells Inc., Seattle, WA, United States
| | - Sergio Hernandez
- Biology Department, California State University Northridge, Northridge, CA, United States
| | - Belle Giang
- Moorpark College, Moorpark, CA, United States
| | - Chris Chabot
- Biology Department, California State University Northridge, Northridge, CA, United States
| | | | - Maria Elena de Bellard
- Biology Department, California State University Northridge, Northridge, CA, United States
| |
Collapse
|
11
|
Pan J, Xu X, Wang G. lncRNA ZFAS1 Is Involved in the Proliferation, Invasion and Metastasis of Prostate Cancer Cells Through Competitively Binding to miR-135a-5p. Cancer Manag Res 2020; 12:1135-1149. [PMID: 32104094 PMCID: PMC7025677 DOI: 10.2147/cmar.s237439] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 01/07/2020] [Indexed: 12/16/2022] Open
Abstract
Background Prostate cancer (PCa) is a common malignant tumor in men. lncRNA ZFAS1 plays a carcinogenic role in many types of cancer; however, its potential role in PCa remains unclear. The current study aimed to determine the expression and function of ZFAS1 in PC. Methods The ZFAS1 expression in PC tissues and cells was determined by quantitative polymerase chain reaction (qPCR). SiZFAS1, miR-135a-5p mimic and miR-135a-5p inhibitor were transfected into PCa cells. The direct target of ZFAS1 was predicted by Starbase and verified by dual-luciferase reporter. Cell viability, proliferation, apoptosis, migration and invasion of the PCa cells were determined by cell counting kit-8, clone formation assay, flow cytometer, scratch and Transwell assay, respectively. The expression levels of related proteins and mRNAs were determined by Western blotting and qPCR. Results ZFAS1 expression was up-regulated in PCa cells and tissues. ZFAS1 could competitively bind to miR-135a-5p in PCa cells, and down-regulation of ZFAS1 inhibited cell viability, proliferation, migration, invasion of PCa cells and the occurrence of epithelial-mesenchymal transformation (EMT) and promoted apoptosis of PCa cells and increased the miR-135a-5p expression. Moreover, the function of miR-135a-5p mimic in PCa cells was consistent with ZFAS1 knockdown, while the function of miR-135a-5p inhibitor was opposite to that of miR-135a-5p mimic in PCa cells. The results showed that knocking down ZFAS1 could attenuate the effects of miR-135a-5p inhibitor on cell proliferation, invasion and EMT of PCa cells. Conclusion Knocking down ZFAS1 could inhibit the proliferation, invasion and metastasis of PCa cells through regulating miR-135a-5p expression.
Collapse
Affiliation(s)
- Jiaqiang Pan
- Department of Urology, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Xingyan Xu
- Department of Ophthalmology, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Guangliang Wang
- Department of Histology and Embryology, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| |
Collapse
|
12
|
Kumar D, Nitzan E, Kalcheim C. YAP promotes neural crest emigration through interactions with BMP and Wnt activities. Cell Commun Signal 2019; 17:69. [PMID: 31228951 PMCID: PMC6589182 DOI: 10.1186/s12964-019-0383-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 06/04/2019] [Indexed: 12/12/2022] Open
Abstract
Background Premigratory neural crest progenitors undergo an epithelial-to-mesenchymal transition and leave the neural tube as motile cells. Previously, we showed that BMP generates trunk neural crest emigration through canonical Wnt signaling which in turn stimulates G1/S transition. The molecular network underlying this process is, however, not yet completely deciphered. Yes-associated-protein (YAP), an effector of the Hippo pathway, controls various aspects of development including cell proliferation, migration, survival and differentiation. In this study, we examined the possible involvement of YAP in neural crest emigration and its relationship with BMP and Wnt. Methods We implemented avian embryos in which levels of YAP gene activity were either reduced or upregulated by in ovo plasmid electroporation, and monitored effects on neural crest emigration, survival and proliferation. Neural crest-derived sensory neuron and melanocyte development were assessed upon gain of YAP function. Imunohistochemistry was used to assess YAP expression. In addition, the activity of specific signaling pathways including YAP, BMP and Wnt was monitored with specific reporters. Results We find that the Hippo pathway transcriptional co-activator YAP is expressed and is active in premigratory crest of avian embryos. Gain of YAP function stimulates neural crest emigration in vivo, and attenuating YAP inhibits cell exit. This is associated with an accumulation of FoxD3-expressing cells in the dorsal neural tube, with reduced proliferation, and enhanced apoptosis. Furthermore, gain of YAP function inhibits differentiation of Islet-1-positive sensory neurons and augments the number of EdnrB2-positive melanocytes. Using specific in vivo reporters, we show that loss of YAP function in the dorsal neural tube inhibits BMP and Wnt activities whereas gain of YAP function stimulates these pathways. Reciprocally, inhibition of BMP and Wnt signaling by noggin or Xdd1, respectively, downregulates YAP activity. In addition, YAP-dependent stimulation of neural crest emigration is compromised upon inhibition of either BMP or Wnt activities. Together, our results suggest a positive bidirectional cross talk between these pathways. Conclusions Our data show that YAP is necessary for emigration of neural crest progenitors. In addition, they incorporate YAP signaling into a BMP/Wnt-dependent molecular network responsible for emigration of trunk-level neural crest.
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, 91120, Jerusalem, Israel
| | - Erez Nitzan
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, 91120, Jerusalem, Israel
| | - Chaya Kalcheim
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, 91120, Jerusalem, Israel.
| |
Collapse
|
13
|
Abstract
Neural crest cells are the embryonic precursors of most neurons and all glia of the peripheral nervous system, pigment cells, some endocrine components, and connective tissue of the head, face, neck, and heart. Following induction, crest cells undergo an epithelial to mesenchymal transition that enables them to migrate along specific pathways culminating in their phenotypic differentiation. Researching this unique embryonic population has revealed important understandings of basic biological and developmental principles. These principles are likely to assist in clarifying the etiology and help in finding strategies for the treatment of neural crest diseases, collectively termed neurocristopathies. The progress achieved in neural crest research is made feasible thanks to the continuous development of species-specific in vivo and in vitro paradigms and more recently the possibility to produce neural crest cells and specific derivatives from embryonic or induced pluripotent stem cells. All of the above assist us in elucidating mechanisms that regulate neural crest development using state-of-the art cellular, molecular, and imaging approaches.
Collapse
Affiliation(s)
- Chaya Kalcheim
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC), Hebrew University-Hadassah Medical School, Jerusalem, Israel.
- Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University-Hadassah Medical School, Jerusalem, Israel.
| |
Collapse
|
14
|
Rogers CD, Nie S. Specifying neural crest cells: From chromatin to morphogens and factors in between. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2018; 7:e322. [PMID: 29722151 PMCID: PMC6215528 DOI: 10.1002/wdev.322] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 03/26/2018] [Accepted: 03/27/2018] [Indexed: 12/16/2022]
Abstract
Neural crest (NC) cells are a stem-like multipotent population of progenitor cells that are present in vertebrate embryos, traveling to various regions in the developing organism. Known as the "fourth germ layer," these cells originate in the ectoderm between the neural plate (NP), which will become the brain and spinal cord, and nonneural tissues that will become the skin and the sensory organs. NC cells can differentiate into more than 30 different derivatives in response to the appropriate signals including, but not limited to, craniofacial bone and cartilage, sensory nerves and ganglia, pigment cells, and connective tissue. The molecular and cellular mechanisms that control the induction and specification of NC cells include epigenetic control, multiple interactive and redundant transcriptional pathways, secreted signaling molecules, and adhesion molecules. NC cells are important not only because they transform into a wide variety of tissue types, but also because their ability to detach from their epithelial neighbors and migrate throughout developing embryos utilizes mechanisms similar to those used by metastatic cancer cells. In this review, we discuss the mechanisms required for the induction and specification of NC cells in various vertebrate species, focusing on the roles of early morphogenesis, cell adhesion, signaling from adjacent tissues, and the massive transcriptional network that controls the formation of these amazing cells. This article is categorized under: Nervous System Development > Vertebrates: General Principles Gene Expression and Transcriptional Hierarchies > Regulatory Mechanisms Gene Expression and Transcriptional Hierarchies > Gene Networks and Genomics Signaling Pathways > Cell Fate Signaling.
Collapse
Affiliation(s)
- Crystal D. Rogers
- Department of Biology, College of Science and Mathematics, California State University Northridge, Northridge, California
| | - Shuyi Nie
- School of Biological Sciences and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
15
|
Latin American contributions to the neural crest field. Mech Dev 2018; 153:17-29. [PMID: 30081090 DOI: 10.1016/j.mod.2018.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 07/15/2018] [Accepted: 07/26/2018] [Indexed: 11/21/2022]
Abstract
The neural crest (NC) is one of the most fascinating structures during embryonic development. Unique to vertebrate embryos, these cells give rise to important components of the craniofacial skeleton, such as the jaws and skull, as well as melanocytes and ganglia of the peripheral nervous system. Worldwide, several groups have been studying NC development and specifically in the Latin America (LA) they have been growing in numbers since the 1990s. It is important for the world to recognize the contributions of LA researchers on the knowledge of NC development, as it can stimulate networking and improvement in the field. We developed a database of LA publications on NC development using ORCID and PUBMED as search engines. We thoroughly describe all of the contributions from LA, collected in five major topics on NC development mechanisms: i) induction and specification; ii) migration; iii) differentiation; iv) adult NC; and, v) neurocristopathies. Further analysis was done to correlate each LA country with topics and animal models, and to access collaboration between LA countries. We observed that some LA countries have made important contributions to the comprehension of NC development. Interestingly, some LA countries have a topic and an animal model as their strength; in addition, collaboration between LA countries is almost inexistent. This review will help LA NC research to be acknowledged, and to facilitate networking between students and researchers worldwide.
Collapse
|
16
|
Kalcheim C. Neural crest emigration: From start to stop. Genesis 2018; 56:e23090. [DOI: 10.1002/dvg.23090] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 01/07/2018] [Accepted: 01/08/2018] [Indexed: 12/28/2022]
Affiliation(s)
- Chaya Kalcheim
- Department of Medical Neurobiology, IMRIC and ELSC; Hebrew University of Jerusalem-Hadassah Medical School; Jerusalem 9112102 Israel
| |
Collapse
|
17
|
Pauli S, Bajpai R, Borchers A. CHARGEd with neural crest defects. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2017; 175:478-486. [PMID: 29082625 DOI: 10.1002/ajmg.c.31584] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 08/23/2017] [Accepted: 08/31/2017] [Indexed: 12/15/2022]
Abstract
Neural crest cells are highly migratory pluripotent cells that give rise to diverse derivatives including cartilage, bone, smooth muscle, pigment, and endocrine cells as well as neurons and glia. Abnormalities in neural crest-derived tissues contribute to the etiology of CHARGE syndrome, a complex malformation disorder that encompasses clinical symptoms like coloboma, heart defects, atresia of the choanae, retarded growth and development, genital hypoplasia, ear anomalies, and deafness. Mutations in the chromodomain helicase DNA-binding protein 7 (CHD7) gene are causative of CHARGE syndrome and loss-of-function data in different model systems have firmly established a role of CHD7 in neural crest development. Here, we will summarize our current understanding of the function of CHD7 in neural crest development and discuss possible links of CHARGE syndrome to other developmental disorders.
Collapse
Affiliation(s)
- Silke Pauli
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Ruchi Bajpai
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry and Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Annette Borchers
- Department of Biology, Molecular Embryology, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
18
|
Nouri M, Caradec J, Lubik AA, Li N, Hollier BG, Takhar M, Altimirano-Dimas M, Chen M, Roshan-Moniri M, Butler M, Lehman M, Bishop J, Truong S, Huang SC, Cochrane D, Cox M, Collins C, Gleave M, Erho N, Alshalafa M, Davicioni E, Nelson C, Gregory-Evans S, Karnes RJ, Jenkins RB, Klein EA, Buttyan R. Therapy-induced developmental reprogramming of prostate cancer cells and acquired therapy resistance. Oncotarget 2017; 8:18949-18967. [PMID: 28145883 PMCID: PMC5386661 DOI: 10.18632/oncotarget.14850] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 01/16/2017] [Indexed: 01/01/2023] Open
Abstract
Treatment-induced neuroendocrine transdifferentiation (NEtD) complicates therapies for metastatic prostate cancer (PCa). Based on evidence that PCa cells can transdifferentiate to other neuroectodermally-derived cell lineages in vitro, we proposed that NEtD requires first an intermediary reprogramming to metastable cancer stem-like cells (CSCs) of a neural class and we demonstrate that several different AR+/PSA+ PCa cell lines were efficiently reprogrammed to, maintained and propagated as CSCs by growth in androgen-free neural/neural crest (N/NC) stem medium. Such reprogrammed cells lost features of prostate differentiation; gained features of N/NC stem cells and tumor-initiating potential; were resistant to androgen signaling inhibition; and acquired an invasive phenotype in vitro and in vivo. When placed back into serum-containing mediums, reprogrammed cells could be re-differentiated to N-/NC-derived cell lineages or return back to an AR+ prostate-like state. Once returned, the AR+ cells were resistant to androgen signaling inhibition. Acute androgen deprivation or anti-androgen treatment in serum-containing medium led to the transient appearance of a sub-population of cells with similar characteristics. Finally, a 132 gene signature derived from reprogrammed PCa cell lines distinguished tumors from PCa patients with adverse outcomes. This model may explain neural manifestations of PCa associated with lethal disease. The metastable nature of the reprogrammed stem-like PCa cells suggests that cycles of PCa cell reprogramming followed by re-differentiation may support disease progression and therapeutic resistance. The ability of a gene signature from reprogrammed PCa cells to identify tumors from patients with metastasis or PCa-specific mortality implies that developmental reprogramming is linked to aggressive tumor behaviors.
Collapse
Affiliation(s)
- Mannan Nouri
- Vancouver Prostate Centre, Vancouver, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Josselin Caradec
- Vancouver Prostate Centre, Vancouver, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Amy Anne Lubik
- Vancouver Prostate Centre, Vancouver, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Na Li
- Vancouver Prostate Centre, Vancouver, Canada
| | - Brett G Hollier
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | | | | | - Mengqian Chen
- Drug Discovery & Biomedical Sciences, South Carolina College of Pharmacy, Columbia, South Carolina, USA
| | | | | | - Melanie Lehman
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | | | | | | | - Dawn Cochrane
- Department of Molecular Oncology, British Columbia Cancer Agency, Vancouver, Canada
| | - Michael Cox
- Vancouver Prostate Centre, Vancouver, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Colin Collins
- Vancouver Prostate Centre, Vancouver, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Martin Gleave
- Vancouver Prostate Centre, Vancouver, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Nicholas Erho
- GenomeDX Biosciences, Vancouver, Canada.,GenomeDX Biosciences, San Diego, California, USA
| | | | - Elai Davicioni
- GenomeDX Biosciences, Vancouver, Canada.,GenomeDX Biosciences, San Diego, California, USA
| | - Colleen Nelson
- Vancouver Prostate Centre, Vancouver, Canada.,Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Sheryl Gregory-Evans
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, Canada
| | | | - Robert B Jenkins
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Eric A Klein
- Glickman Urological and Kidney Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Ralph Buttyan
- Vancouver Prostate Centre, Vancouver, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| |
Collapse
|
19
|
Cadherin-11 promotes neural crest cell spreading by reducing intracellular tension-Mapping adhesion and mechanics in neural crest explants by atomic force microscopy. Semin Cell Dev Biol 2017; 73:95-106. [PMID: 28919310 DOI: 10.1016/j.semcdb.2017.08.058] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 08/24/2017] [Accepted: 08/28/2017] [Indexed: 11/21/2022]
Abstract
During development cranial neural crest cells (NCCs) display a striking transition from collective to single-cell migration, but the mechanisms enabling individual NCCs to separate from the neural crest tissue are still incompletely understood. In this study we have employed atomic force microscopy (AFM) to investigate potential adhesive and mechanical changes associated with the dissociation of individual cells from cohesive Xenopus NCC explants at early stages of migration. AFM-based single-cell force spectroscopy (SCFS) revealed a uniform distribution of cell-cell adhesion forces within NCC explants, including semi-detached leader cells in the process of delaminating from the explant edge. This suggested that dissociation from the cell sheet may not require prior weakening of cell-cell contacts. However, mapping NCC sheet elasticity by AFM microbead indentation demonstrated strongly reduced cell stiffness in semi-detached leader cells compared to neighbouring cells in the NCC sheet periphery. Reduced leader cell stiffness coincided with enhanced cell spreading and high substrate traction, indicating a possible mechano-regulation of leader cell delamination. In support, AFM elasticity measurements of individual NCCs in optical side view mode demonstrated that reducing cell tension by inhibiting actomyosin contractility induces rapid spreading, possibly maximizing cell-substrate interactions as a result. Depletion of cadherin-11, a classical cadherin with an essential role in NCC migration and substrate adhesion, prevented the tension reduction necessary for NCC spreading, both in individual cells and at the edge of explanted sheets. In contrast, overexpression of cadherin-11 accelerated spreading of both individual cells and delaminating leader cells. As cadherin-11 expression increases strongly during NCC migration, this suggests an important role of cadherin-11 in regulating NCC elasticity and spreading at later stages of NCC migration. We therefore propose a model in which high tension at the NCC sheet periphery prevents premature NCC spreading and delamination during early stages of migration, while a cadherin-11-dependent local decrease in cell tension promotes leader cell spreading and delamination at later stages of migration.
Collapse
|
20
|
York JR, Yuan T, Zehnder K, McCauley DW. Lamprey neural crest migration is Snail-dependent and occurs without a differential shift in cadherin expression. Dev Biol 2017. [PMID: 28624345 DOI: 10.1016/j.ydbio.2017.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The acquisition of neural crest cells was a key step in the origin of the vertebrate body plan. An outstanding question is how neural crest cells acquired their ability to undergo an epithelial-mesenchymal transition (EMT) and migrate extensively throughout the vertebrate embryo. We tested if differential regulation of classical cadherins-a highly conserved feature of neural crest EMT and migration in jawed vertebrates-mediates these cellular behaviors in lamprey, a basal jawless vertebrate. Lamprey has single copies of the type I and type II classical cadherins (CadIA and CadIIA). CadIIA is expressed in premigratory neural crest, and requires the transcription factor Snail for proper expression, yet CadIA is never expressed in the neural tube during neural crest development, suggesting that differential regulation of classical cadherin expression is not required to initiate neural crest migration in basal vertebrates. We hypothesize that neural crest cells evolved by retention of regulatory programs linking distinct mesenchymal and multipotency properties, and emigrated from the neural tube without differentially regulating type I/type II cadherins. Our results point to the coupling of mesenchymal state and multipotency as a key event facilitating the origin of migratory neural crest cells.
Collapse
Affiliation(s)
- Joshua R York
- Department of Biology, University of Oklahoma, 730 Van Vleet Oval, Norman, OK 73019, USA
| | - Tian Yuan
- Department of Biology, University of Oklahoma, 730 Van Vleet Oval, Norman, OK 73019, USA
| | - Kevin Zehnder
- Department of Biology, University of Oklahoma, 730 Van Vleet Oval, Norman, OK 73019, USA
| | - David W McCauley
- Department of Biology, University of Oklahoma, 730 Van Vleet Oval, Norman, OK 73019, USA.
| |
Collapse
|
21
|
Dady A, Duband JL. Cadherin interplay during neural crest segregation from the non-neural ectoderm and neural tube in the early chick embryo. Dev Dyn 2017; 246:550-565. [PMID: 28474787 DOI: 10.1002/dvdy.24517] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 04/19/2017] [Accepted: 04/26/2017] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND In the avian embryo, neural crest (NC) progenitors arise in the neuroectoderm during gastrulation, long before their dissemination. Although the gene regulatory network involved in NC specification has been deciphered, the mechanisms involved in their segregation from the other neuroectoderm-derived progenitors, notably the epidermis and neural tube, are unknown. Because cadherins mediate cell recognition and sorting, we scrutinized their expression profiles during NC specification and delamination. RESULTS We found that the NC territory is defined precociously by the robust expression of Cadherin-6B in cells initially scattered among other cells uniformly expressing E-cadherin, and that NC progenitors are progressively sorted and regrouped into a discrete domain between the prospective epidermis and neural tube. At completion of NC specification, the epidermis, NC, and neural tube are fully segregated in contiguous compartments characterized by distinct cadherin repertoires. We also found that Cadherin-6B down-regulation constitutes a major event during NC delamination and that, with the exception of the caudal part of the embryo, N-cadherin is unlikely to control NC emigration. CONCLUSIONS Our results indicate that partition of the neuroectoderm is mediated by cadherin interplays and ascribes a key role to Cadherin-6B in the specification and delamination of the NC population. Developmental Dynamics 246:550-565, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Alwyn Dady
- Laboratoire de Biologie du Développement, Centre National de la Recherche Scientifique, Paris, France.,Laboratoire de Biologie du Développement, Université Pierre et Marie Curie, Paris, France
| | - Jean-Loup Duband
- Laboratoire de Biologie du Développement, Centre National de la Recherche Scientifique, Paris, France.,Laboratoire de Biologie du Développement, Université Pierre et Marie Curie, Paris, France.,Institut Mondor de Recherches Biomédicales, Institut National de la Santé et de la Recherche Médicale, Créteil, France.,Institut Mondor de Recherches Biomédicales, Université Paris-Est Créteil, Créteil, France
| |
Collapse
|
22
|
Li J, Qu J, Shi Y, Perfetto M, Ping Z, Christian L, Niu H, Mei S, Zhang Q, Yang X, Wei S. Nicotinic acid inhibits glioma invasion by facilitating Snail1 degradation. Sci Rep 2017; 7:43173. [PMID: 28256591 PMCID: PMC5335718 DOI: 10.1038/srep43173] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 01/23/2017] [Indexed: 01/22/2023] Open
Abstract
Malignant glioma is a formidable disease that commonly leads to death, mainly due to the invasion of tumor cells into neighboring tissues. Therefore, inhibition of tumor cell invasion may provide an effective therapy for malignant glioma. Here we report that nicotinic acid (NA), an essential vitamin, inhibits glioma cell invasion in vitro and in vivo. Treatment of the U251 glioma cells with NA in vitro results in reduced invasion, which is accompanied by a loss of mesenchymal phenotype and an increase in cell-cell adhesion. At the molecular level, transcription of the adherens junction protein E-cadherin is upregulated, leading to accumulation of E-cadherin protein at the cell-cell boundary. This can be attributed to NA's ability to facilitate the ubiquitination and degradation of Snail1, a transcription factor that represses E-cadherin expression. Similarly, NA transiently inhibits neural crest migration in Xenopus embryos in a Snail1-dependent manner, indicating that the mechanism of action for NA in cell migration is evolutionarily conserved. We further show that NA injection blocks the infiltration of tumor cells into the adjacent brain tissues and improves animal survival in a rat model of glioma. These results suggest that NA treatment may be developed into a potential therapy for malignant glioma.
Collapse
Affiliation(s)
- Jiejing Li
- Department of Clinical Laboratory, The Affiliated Hospital of KMUST, Medical School, Kunming University of Science and Technology, Kunming 650032, China.,Department of Biology, West Virginia University, Morgantown, WV 26506, United States
| | - Jiagui Qu
- Department of Clinical Laboratory, The Affiliated Hospital of KMUST, Medical School, Kunming University of Science and Technology, Kunming 650032, China
| | - Yu Shi
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, Chongqing 400014, China.,Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics; Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing 400014, China
| | - Mark Perfetto
- Department of Biology, West Virginia University, Morgantown, WV 26506, United States.,Department of Biological Sciences, University of Delaware, Newark, DE 19716, United States
| | - Zhuxian Ping
- Department of Clinical Laboratory, The Affiliated Hospital of KMUST, Medical School, Kunming University of Science and Technology, Kunming 650032, China
| | - Laura Christian
- Department of Biology, West Virginia University, Morgantown, WV 26506, United States
| | - Hua Niu
- Department of Clinical Laboratory, The Affiliated Hospital of KMUST, Medical School, Kunming University of Science and Technology, Kunming 650032, China
| | - Shuting Mei
- Department of Gerontology, First People's Hospital of Yunnan Province, Kunming 650032, China
| | - Qin Zhang
- Department of Clinical Laboratory, The Affiliated Hospital of KMUST, Medical School, Kunming University of Science and Technology, Kunming 650032, China
| | - Xiangcai Yang
- Department of Clinical Laboratory, The Affiliated Hospital of KMUST, Medical School, Kunming University of Science and Technology, Kunming 650032, China
| | - Shuo Wei
- Department of Biology, West Virginia University, Morgantown, WV 26506, United States.,Department of Biological Sciences, University of Delaware, Newark, DE 19716, United States
| |
Collapse
|
23
|
Epithelial-mesenchymal transition in morphogenesis, cancer progression and angiogenesis. Exp Cell Res 2017; 353:1-5. [PMID: 28257786 DOI: 10.1016/j.yexcr.2017.02.041] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 02/22/2017] [Accepted: 02/27/2017] [Indexed: 12/18/2022]
Abstract
All organs consist of an epithelium and an associated mesenchyme, so these epithelial-mesenchymal intercations are among the most important phenomena in nature. The aim of this article is the summarize the common mechanisms involved in the establishment of epithelial mesenchymal transition in three biological processes, namely organogenesis, tumor progression and metastasis, and angiogenesis, apparently independent each from other. A common feature of these processes is the fact that specialized epithelial cells lose their features, including cell adhesion and polarity, reorganize their cytoskeleton, and acquire a mesenchymal morphology and the ability to migrate.
Collapse
|
24
|
Zage PE, Whittle SB, Shohet JM. CD114: A New Member of the Neural Crest-Derived Cancer Stem Cell Marker Family. J Cell Biochem 2016; 118:221-231. [PMID: 27428599 DOI: 10.1002/jcb.25656] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 07/15/2016] [Indexed: 12/13/2022]
Abstract
The neural crest is a population of cells in the vertebrate embryo that gives rise to a wide range of tissues and cell types, including components of the peripheral nervous system and the craniofacial skeleton as well as melanocytes and the adrenal medulla. Aberrations in neural crest development can lead to numerous diseases, including cancers such as melanoma and neuroblastoma. Cancer stem cells (CSCs) have been identified in these neural crest-derived tumors, and these CSCs demonstrate resistance to treatment and are likely key contributors to disease relapse. Patients with neural crest-derived tumors often have poor outcomes due to frequent relapses, likely due to the continued presence of residual treatment-resistant CSCs, and therapies directed against these CSCs are likely to improve patient outcomes. CSCs share many of the same genetic and biologic features of primordial neural crest cells, and therefore a better understanding of neural crest development will likely lead to the development of effective therapies directed against these CSCs. Signaling through STAT3 has been shown to be required for neural crest development, and granulocyte colony stimulating factor (GCSF)-mediated activation of STAT3 has been shown to play a role in the pathogenesis of neural crest-derived tumors. Expression of the cell surface marker CD114 (the receptor for GCSF) has been identified as a potential marker for CSCs in neural crest-derived tumors, suggesting that CD114 expression and function may contribute to disease relapse and poor patient outcomes. Here we review the processes of neural crest development and tumorigenesis and we discuss the previously identified markers for CSC subpopulations identified in neural crest tumors and their role in neural crest tumor biology. We also discuss the potential for CD114 and downstream intracellular signaling pathways as potential targets for CSC-directed therapy. J. Cell. Biochem. 118: 221-231, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Peter E Zage
- Division of Hematology-Oncology, Department of Pediatrics, University of California San Diego, La Jolla, California.,Peckham Center for Cancer and Blood Disorders, Rady Children's Hospital, San Diego, California
| | - Sarah B Whittle
- Department of Pediatrics, Section of Hematology-Oncology, Children's Cancer Center, Houston, Texas
| | - Jason M Shohet
- Department of Pediatrics, Section of Hematology-Oncology, Children's Cancer Center, Houston, Texas.,Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
25
|
Wilson NR, Olm-Shipman AJ, Acevedo DS, Palaniyandi K, Hall EG, Kosa E, Stumpff KM, Smith GJ, Pitstick L, Liao EC, Bjork BC, Czirok A, Saadi I. SPECC1L deficiency results in increased adherens junction stability and reduced cranial neural crest cell delamination. Sci Rep 2016; 6:17735. [PMID: 26787558 PMCID: PMC4726231 DOI: 10.1038/srep17735] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 11/05/2015] [Indexed: 11/16/2022] Open
Abstract
Cranial neural crest cells (CNCCs) delaminate from embryonic neural folds and migrate to pharyngeal arches, which give rise to most mid-facial structures. CNCC dysfunction plays a prominent role in the etiology of orofacial clefts, a frequent birth malformation. Heterozygous mutations in SPECC1L have been identified in patients with atypical and syndromic clefts. Here, we report that in SPECC1L-knockdown cultured cells, staining of canonical adherens junction (AJ) components, β-catenin and E-cadherin, was increased, and electron micrographs revealed an apico-basal diffusion of AJs. To understand the role of SPECC1L in craniofacial morphogenesis, we generated a mouse model of Specc1l deficiency. Homozygous mutants were embryonic lethal and showed impaired neural tube closure and CNCC delamination. Staining of AJ proteins was increased in the mutant neural folds. This AJ defect is consistent with impaired CNCC delamination, which requires AJ dissolution. Further, PI3K-AKT signaling was reduced and apoptosis was increased in Specc1l mutants. In vitro, moderate inhibition of PI3K-AKT signaling in wildtype cells was sufficient to cause AJ alterations. Importantly, AJ changes induced by SPECC1L-knockdown were rescued by activating the PI3K-AKT pathway. Together, these data indicate SPECC1L as a novel modulator of PI3K-AKT signaling and AJ biology, required for neural tube closure and CNCC delamination.
Collapse
Affiliation(s)
- Nathan R Wilson
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Adam J Olm-Shipman
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Diana S Acevedo
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kanagaraj Palaniyandi
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Everett G Hall
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Edina Kosa
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kelly M Stumpff
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Guerin J Smith
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Lenore Pitstick
- Department of Biochemistry, Midwestern University, Downers Grove, IL, USA
| | - Eric C Liao
- Center for Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Bryan C Bjork
- Department of Biochemistry, Midwestern University, Downers Grove, IL, USA
| | - Andras Czirok
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Irfan Saadi
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
26
|
Bottoni P, Isgrò MA, Scatena R. The epithelial-mesenchymal transition in cancer: a potential critical topic for translational proteomic research. Expert Rev Proteomics 2015; 13:115-33. [PMID: 26567562 DOI: 10.1586/14789450.2016.1112742] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The epithelial-mesenchymal transition (EMT) is a morphogenetic process that results in a loss of epithelial characteristics and the acquisition of a mesenchymal phenotype. First described in embryogenesis, the EMT has been recently implicated in carcinogenesis and tumor progression. In addition, recent evidence has shown that stem-like cancer cells present the hallmarks of the EMT. Some of the molecular mechanisms related to the interrelationships between cancer pathophysiology and the EMT are well-defined. Nevertheless, the precise molecular mechanism by which epithelial cancer cells acquire the mesenchymal phenotype remains largely unknown. This review focuses on various proteomic strategies with the goal of better understanding the physiological and pathological mechanisms of the EMT process.
Collapse
Affiliation(s)
- Patrizia Bottoni
- a Institute of Biochemistry and Clinical Biochemistry , School of Medicine - Catholic University , Rome , Italy
| | - Maria Antonietta Isgrò
- b Department of Diagnostic and Molecular Medicine , Catholic University of the Sacred Heart , Rome , Italy
| | - Roberto Scatena
- a Institute of Biochemistry and Clinical Biochemistry , School of Medicine - Catholic University , Rome , Italy
| |
Collapse
|
27
|
Kuo CS, Krasnow MA. Formation of a Neurosensory Organ by Epithelial Cell Slithering. Cell 2015; 163:394-405. [PMID: 26435104 DOI: 10.1016/j.cell.2015.09.021] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 07/08/2015] [Accepted: 08/11/2015] [Indexed: 11/25/2022]
Abstract
Epithelial cells are normally stably anchored, maintaining their relative positions and association with the basement membrane. Developmental rearrangements occur through cell intercalation, and cells can delaminate during epithelial-mesenchymal transitions and metastasis. We mapped the formation of lung neuroepithelial bodies (NEBs), innervated clusters of neuroendocrine/neurosensory cells within the bronchial epithelium, revealing a targeted mode of cell migration that we named "slithering," in which cells transiently lose epithelial character but remain associated with the membrane while traversing neighboring epithelial cells to reach cluster sites. Immunostaining, lineage tracing, clonal analysis, and live imaging showed that NEB progenitors, initially distributed randomly, downregulate adhesion and polarity proteins, crawling over and between neighboring cells to converge at diametrically opposed positions at bronchial branchpoints, where they reestablish epithelial structure and express neuroendocrine genes. There is little accompanying progenitor proliferation or apoptosis. Activation of the slithering program may explain why lung cancers arising from neuroendocrine cells are highly metastatic.
Collapse
Affiliation(s)
- Christin S Kuo
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305-5307, USA; Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305-5307, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305-5307, USA
| | - Mark A Krasnow
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305-5307, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305-5307, USA.
| |
Collapse
|
28
|
Agarwal S, Lakoma A, Chen Z, Hicks J, Metelitsa LS, Kim ES, Shohet JM. G-CSF Promotes Neuroblastoma Tumorigenicity and Metastasis via STAT3-Dependent Cancer Stem Cell Activation. Cancer Res 2015; 75:2566-79. [PMID: 25908586 DOI: 10.1158/0008-5472.can-14-2946] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 04/11/2015] [Indexed: 12/16/2022]
Abstract
Increasing evidence suggests that inflammatory cytokines play a critical role in tumor initiation and progression. A cancer stem cell (CSC)-like subpopulation in neuroblastoma is known to be marked by expression of the G-CSF receptor (G-CSFR). Here, we report on the mechanistic contributions of the G-CSFR in neuroblastoma CSCs. Specifically, we demonstrate that the receptor ligand G-CSF selectively activates STAT3 within neuroblastoma CSC subpopulations, promoting their expansion in vitro and in vivo. Exogenous G-CSF enhances tumor growth and metastasis in human xenograft and murine neuroblastoma tumor models. In response to G-CSF, STAT3 acts in a feed-forward loop to transcriptionally activate the G-CSFR and sustain neuroblastoma CSCs. Blockade of this G-CSF-STAT3 signaling loop with either anti-G-CSF antibody or STAT3 inhibitor depleted the CSC subpopulation within tumors, driving correlated tumor growth inhibition, decreased metastasis, and increased chemosensitivity. Taken together, our results define G-CSF as a CSC-activating factor in neuroblastoma, suggest a comprehensive reevaluation of the clinical use of G-CSF in these patients to support white blood cell counts, and suggest that direct targeting of the G-CSF-STAT3 signaling represents a novel therapeutic approach for neuroblastoma.
Collapse
Affiliation(s)
- Saurabh Agarwal
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer Center, Houston, Texas. Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Anna Lakoma
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Zaowen Chen
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer Center, Houston, Texas. Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - John Hicks
- Department of Pathology, Section of Pediatric Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Leonid S Metelitsa
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer Center, Houston, Texas. Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Eugene S Kim
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas. Division of Pediatric Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, California
| | - Jason M Shohet
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer Center, Houston, Texas. Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
29
|
Barriga EH, Mayor R. Embryonic cell-cell adhesion: a key player in collective neural crest migration. Curr Top Dev Biol 2015; 112:301-23. [PMID: 25733144 DOI: 10.1016/bs.ctdb.2014.11.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell migration is essential for morphogenesis, adult tissue remodeling, wound healing, and cancer cell migration. Cells can migrate as individuals or groups. When cells migrate in groups, cell-cell interactions are crucial in order to promote the coordinated behavior, essential for collective migration. Interestingly, recent evidence has shown that cell-cell interactions are also important for establishing and maintaining the directionality of these migratory events. We focus on neural crest cells, as they possess extraordinary migratory capabilities that allow them to migrate and colonize tissues all over the embryo. Neural crest cells undergo an epithelial-to-mesenchymal transition at the same time than perform directional collective migration. Cell-cell adhesion has been shown to be an important source of planar cell polarity and cell coordination during collective movement. We also review molecular mechanisms underlying cadherin turnover, showing how the modulation and dynamics of cell-cell adhesions are crucial in order to maintain tissue integrity and collective migration in vivo. We conclude that cell-cell adhesion during embryo development cannot be considered as simple passive resistance to force, but rather participates in signaling events that determine important cell behaviors required for cell migration.
Collapse
Affiliation(s)
- Elias H Barriga
- Cell and Developmental Biology Department, University College London, London, United Kingdom
| | - Roberto Mayor
- Cell and Developmental Biology Department, University College London, London, United Kingdom.
| |
Collapse
|
30
|
Duband JL, Dady A, Fleury V. Resolving time and space constraints during neural crest formation and delamination. Curr Top Dev Biol 2015; 111:27-67. [PMID: 25662257 DOI: 10.1016/bs.ctdb.2014.11.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A striking feature of neural crest development in vertebrates is that all the specification, delamination, migration, and differentiation steps occur consecutively in distinct areas of the embryo and at different timings of development. The significance and consequences of this partition into clearly separated events are not fully understood yet, but it ought to be related to the necessity of controlling precisely and independently each step, given the wide array of cell types and tissues derived from the neural crest and the long duration of their development spanning almost the entire embryonic life. In this chapter, using the examples of early neural crest induction and delamination, we discuss how time and space constraints influence their development and describe the molecular and cellular responses that are employed by cells to adapt. In the first example, we analyze how cell sorting and cell movements cooperate to allow nascent neural crest cells, which are initially mingled with other neurectodermal progenitors after induction, to segregate from the neural tube and ectoderm populations and settle at the apex of the neural tube prior to migration. In the second example, we examine how cadherins drive the entire process of neural crest segregation from the rest of the neurectoderm by their dual role in mediating first cell sorting and cohesion during specification and later in promoting their delamination. In the third example, we describe how the expression and activity of the transcription factors known to drive epithelium-to-mesenchyme transition (EMT) are regulated timely and spatially by the cellular machinery so that they can alternatively and successively regulate neural crest specification and delamination. In the last example, we briefly tackle the problem of how factors triggering EMT may elicit different cell responses in neural tube and neural crest progenitors.
Collapse
Affiliation(s)
- Jean-Loup Duband
- Laboratoire de Biologie du Développement, Université Pierre et Marie Curie-Paris 6, Paris, France; CNRS, Laboratoire de Biologie du Développement, Paris, France.
| | - Alwyn Dady
- Laboratoire de Biologie du Développement, Université Pierre et Marie Curie-Paris 6, Paris, France; CNRS, Laboratoire de Biologie du Développement, Paris, France
| | - Vincent Fleury
- Laboratoire Matière et Systèmes Complexes, CNRS et Université Denis-Diderot-Paris 7, Paris, France
| |
Collapse
|
31
|
Franco C, Hess S. Recent proteomic advances in developmental, regeneration, and cancer governing signaling pathways. Proteomics 2014; 15:1014-25. [PMID: 25316175 DOI: 10.1002/pmic.201400368] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/16/2014] [Accepted: 10/09/2014] [Indexed: 12/12/2022]
Abstract
Embryonic development, adult tissue repair, and cancer share a number of common regulating pathways. The basic processes that govern the events that induce mesenchymal properties in epithelial cells-a process known as epithelial-mesenchymal transition-are central for embryonic development, and can be resumed in adults either during wound healing or tissue regeneration. A misregulation of these pathways is involved in pathological situations, such as tissue fibrosis and cancer. Proteomic approaches have emerged as promising tools to better understand the signaling pathways that govern these complex biological processes. This review focuses on the recent proteomic-based contributions to better understand the modulation of transforming growth factor-beta (TGF-β), wingless-type MMTV integration site family (Wnt), Notch and Receptor tyrosine kinase (RTK) signaling pathways. New advances include the description of new protein interactions, the formation of new protein complexes or the description on how some PTMs are regulating these pathways. Understanding protein interactions and the tempo-spatial modulation of these pathways might lead us to interesting research quests in cancer, embryonic development or even on improving adult tissue regeneration capabilities.
Collapse
Affiliation(s)
- Catarina Franco
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | | |
Collapse
|
32
|
Breau MA, Schneider-Maunoury S. Cranial placodes: models for exploring the multi-facets of cell adhesion in epithelial rearrangement, collective migration and neuronal movements. Dev Biol 2014; 401:25-36. [PMID: 25541234 DOI: 10.1016/j.ydbio.2014.12.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 12/08/2014] [Accepted: 12/09/2014] [Indexed: 01/16/2023]
Abstract
Key to morphogenesis is the orchestration of cell movements in the embryo, which requires fine-tuned adhesive interactions between cells and their close environment. The neural crest paradigm has provided important insights into how adhesion dynamics control epithelium-to-mesenchyme transition and mesenchymal cell migration. Much less is known about cranial placodes, patches of ectodermal cells that generate essential parts of vertebrate sensory organs and ganglia. In this review, we summarise the known functions of adhesion molecules in cranial placode morphogenesis, and discuss potential novel implications of adhesive interactions in this crucial developmental process. The great repertoire of placodal cell behaviours offers new avenues for exploring the multiple roles of adhesion complexes in epithelial remodelling, collective migration and neuronal movements.
Collapse
Affiliation(s)
- Marie Anne Breau
- Sorbonne Universités, UPMC Univ Paris 06, IBPS-UMR7622, F-75005 Paris, France; CNRS, UMR7622, Institut de Biologie Paris-Seine (IBPS) - Laboratoire de Biologie du Développement, F-75005 Paris, France; INSERM, U1156, F-75005 Paris, France.
| | - Sylvie Schneider-Maunoury
- Sorbonne Universités, UPMC Univ Paris 06, IBPS-UMR7622, F-75005 Paris, France; CNRS, UMR7622, Institut de Biologie Paris-Seine (IBPS) - Laboratoire de Biologie du Développement, F-75005 Paris, France; INSERM, U1156, F-75005 Paris, France
| |
Collapse
|
33
|
Hu N, Strobl-Mazzulla PH, Simoes-Costa M, Sánchez-Vásquez E, Bronner ME. DNA methyltransferase 3B regulates duration of neural crest production via repression of Sox10. Proc Natl Acad Sci U S A 2014; 111:17911-6. [PMID: 25453070 PMCID: PMC4273375 DOI: 10.1073/pnas.1318408111] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Neural crest stem cells arise within the central nervous system but then undergo an epithelial-to-mesenchymal transition to migrate away and contribute to the peripheral nervous system and craniofacial skeleton. Here we show that DNA methyltransferase 3B (DNMT3B) is responsible for the loss of competence of dorsal neural tube cells to generate emigrating neural crest cells. DNMT3B knockdown results in up-regulation of neural crest markers, prolonged neural crest emigration, and subsequent precocious neuronal differentiation of the trigeminal ganglion. We find that DNMT3B binds to the promoter of Sox10, known to be important for neural crest emigration and lineage acquisition. Bisulfite sequencing further reveals methylation of the Sox10 promoter region upon cessation of emigration in normal embryos, whereas this mark is reduced after DNMT3B loss. Taken together, these results reveal the importance of DNA methylation in regulating the ability of neural tube cells to produce neural crest cells and the timing of peripheral neuron differentiation.
Collapse
Affiliation(s)
- Na Hu
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125 and
| | - Pablo H Strobl-Mazzulla
- Laboratorio de biología del desarrollo, Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús (CONICET-UNSAM), Chascomús, 7130 Buenos Aires, Argentina
| | - Marcos Simoes-Costa
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125 and
| | - Estefania Sánchez-Vásquez
- Laboratorio de biología del desarrollo, Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús (CONICET-UNSAM), Chascomús, 7130 Buenos Aires, Argentina
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125 and
| |
Collapse
|
34
|
Chen P, Chen JZ, Shao CY, Li CY, Zhang YD, Lu WJ, Fu Y, Gu P, Fan X. Treatment with retinoic acid and lens epithelial cell-conditioned medium in vitro directed the differentiation of pluripotent stem cells towards corneal endothelial cell-like cells. Exp Ther Med 2014; 9:351-360. [PMID: 25574197 PMCID: PMC4280952 DOI: 10.3892/etm.2014.2103] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 11/07/2014] [Indexed: 12/13/2022] Open
Abstract
Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have extensive self-renewal capacity and the potential to differentiate into all tissue-specific cell lineages, including corneal endothelial cells (CECs). They are a promising prospect for the future of regenerative medicine. The method of derivation of CECs from ESCs and iPSCs, however, remains to be elucidated. In this study, mouse ESCs and iPSCs were induced to differentiate into CECs using CEC embryonic development events as a guide. All-trans retinoic acid (RA) treatment during the embryoid body (EB) differentiation step was used to promote neural crest (NC) cell differentiation as first step and was followed by a second induction in CEC- or lens epithelial cell (LEC)-conditioned medium (CM) to ultimately generate CEC-like cells. During the corresponding differentiation stages, NC developmental markers and CEC differentiation markers were detected at the protein level using immunocytochemistry (ICC) and at the mRNA level by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). During the first stage, the data indicated that 4 days of treatment with 1 μM RA starting on day 4 of EB formation favored NC cell differentiation and that plating on gelatin-coated plates led to cell migration out of the EBs. The second-stage differentiation results showed that the CM, particularly the LEC-CM, enhanced the yield of polygonal cells with CEC-specific marker expression shown by ICC and RT-qPCR. This study demonstrates that mouse ESCs and iPSCs were induced and expressed CEC differentiation markers when subjected to a two-step inducement process, suggesting that they are a promising resource for corneal endothelium failure replacement therapy in the future.
Collapse
Affiliation(s)
- Ping Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China ; Department of Ophthalmology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - Jun-Zhao Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - Chun-Yi Shao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - Chuan-Yin Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - Yi-Dan Zhang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - Wen-Juan Lu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - Yao Fu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - Ping Gu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
35
|
Abstract
Neuroblastoma is a developmental tumor of young children arising from the embryonic sympathoadrenal lineage of the neural crest. Neuroblastoma is the primary cause of death from pediatric cancer for children between the ages of one and five years and accounts for ∼13% of all pediatric cancer mortality. Its clinical impact and unique biology have made this aggressive malignancy the focus of a large concerted translational research effort. New insights into tumor biology are driving the development of new classification schemas. Novel targeted therapeutic approaches include small-molecule inhibitors as well as epigenetic, noncoding-RNA, and cell-based immunologic therapies. In this review, recent insights regarding the pathogenesis and biology of neuroblastoma are placed in context with the current understanding of tumor biology and tumor/host interactions. Systematic classification of patients coupled with therapeutic advances point to a future of improved clinical outcomes for this biologically distinct and highly aggressive pediatric malignancy.
Collapse
Affiliation(s)
- Chrystal U Louis
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas 77030; ,
| | | |
Collapse
|
36
|
Cell jamming: collective invasion of mesenchymal tumor cells imposed by tissue confinement. Biochim Biophys Acta Gen Subj 2014; 1840:2386-95. [PMID: 24721714 DOI: 10.1016/j.bbagen.2014.03.020] [Citation(s) in RCA: 208] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 03/25/2014] [Accepted: 03/28/2014] [Indexed: 11/20/2022]
Abstract
BACKGROUND Cancer invasion is a multi-step process which coordinates interactions between tumor cells with mechanotransduction towards the surrounding matrix, resulting in distinct cancer invasion strategies. Defined by context, mesenchymal tumors, including melanoma and fibrosarcoma, develop either single-cell or collective invasion modes, however, the mechanical and molecular programs underlying such plasticity of mesenchymal invasion programs remain unclear. METHODS To test how tissue anatomy determines invasion mode, spheroids of MV3 melanoma and HT1080 fibrosarcoma cells were embedded into 3D collagen matrices of varying density and stiffness and analyzed for migration type and efficacy with matrix metalloproteinase (MMP)-dependent collagen degradation enabled or pharmacologically inhibited. RESULTS With increasing collagen density and dependent on proteolytic collagen breakdown and track clearance, but independent of matrix stiffness, cells switched from single-cell to collective invasion modes. Conversion to collective invasion included gain of cell-to-cell junctions, supracellular polarization and joint guidance along migration tracks. CONCLUSIONS The density of the extracellulair matrix (ECM) determines the invasion mode of mesenchymal tumor cells. Whereas fibrillar, high porosity ECM enables single-cell dissemination, dense matrix induces cell-cell interaction, leader-follower cell behavior and collective migration as an obligate protease-dependent process. GENERAL SIGNIFICANCE These findings establish plasticity of cancer invasion programs in response to ECM porosity and confinement, thereby recapitulating invasion patterns of mesenchymal tumors in vivo. The conversion to collective invasion with increasing ECM confinement supports the concept of cell jamming as a guiding principle for melanoma and fibrosarcoma cells into dense tissue. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
|
37
|
Fort P, Théveneau E. PleiotRHOpic: Rho pathways are essential for all stages of Neural Crest development. Small GTPases 2014; 5:e27975. [PMID: 24614304 DOI: 10.4161/sgtp.27975] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Neural Crest (NC) cells are a multipotent migratory stem cell population unique to vertebrates, which contributes extensively to the formation of a wide array of neural and non-neural structures in the embryo. NC cells originate in the ectoderm at the border of the neural tube, undergo an epithelial-mesenchymal transition and acquire outstanding individual and collective migratory properties that allow them to disseminate and differentiate to different parts of the body. This exquisite capacity to switch from an epithelium to motile cells represents both a puzzling biological issue and an attractive model to address the basic mechanisms of cell migration and their alteration during cancer progression. Here we review how signaling pathways controlled by Rho GTPases, key players in cell adhesion, contraction, migration and polarity, contribute to the control the different phases of NC development.
Collapse
Affiliation(s)
- Philippe Fort
- CNRS; University Montpellier 2; CRBM-UMR5237; Montpellier, France
| | - Eric Théveneau
- CNRS; University Toulouse III; Centre de Biologie du Développement; UMR5547; Toulouse, France
| |
Collapse
|
38
|
The role of the non-canonical Wnt-planar cell polarity pathway in neural crest migration. Biochem J 2014; 457:19-26. [PMID: 24325550 DOI: 10.1042/bj20131182] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The neural crest is an embryonic stem cell population whose migratory behaviour has been likened to malignant invasion. The neural crest, as does cancer, undergoes an epithelial-to-mesenchymal transition and migrates to colonize almost all the tissues of the embryo. Neural crest cells exhibit collective cell migration, moving in streams of high directionality. The migratory neural crest streams are kept in shape by the presence of negative signals in their vicinity. The directionality of the migrating neural crest is achieved by contact-dependent cell polarization, in a phenomenon called contact inhibition of locomotion. Two cells experiencing contact inhibition of locomotion move away from each other after collision. However, if the cell density is high only cells exposed to a free edge can migrate away from the cluster leading to the directional migration of the whole group. Recent work performed in chicks, zebrafish and frogs has shown that the non-canonical Wnt-PCP (planar cell polarity) pathway plays a major role in neural crest migration. PCP signalling controls contact inhibition of locomotion between neural crest cells by localizing different PCP proteins at the site of cell contact during collision and locally regulating the activity of Rho GTPases. Upon collision RhoA (ras homologue family member A) is activated, whereas Rac1 is inhibited at the contact between two migrating neural crest cells, leading to the collapse of protrusions and the migration of cells away from one another. The present review summarizes the mechanisms that control neural crest migration and focuses on the role of non-canonical Wnt or PCP signalling in this process.
Collapse
|
39
|
Czarnobaj J, Bagnall KM, Bamforth JS, Milos NC. The different effects on cranial and trunk neural crest cell behaviour following exposure to a low concentration of alcohol in vitro. Arch Oral Biol 2014; 59:500-12. [PMID: 24631632 DOI: 10.1016/j.archoralbio.2014.02.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 12/17/2013] [Accepted: 02/10/2014] [Indexed: 11/19/2022]
Abstract
Embryonic neural crest cells give rise to large regions of the face and peripheral nervous system. Exposure of these cells to high alcohol concentrations leads to cell death in the craniofacial region resulting in facial defects. However, the effects of low concentrations of alcohol on neural crest cells are not clear. In this study, cranial neural crest cells from Xenopus laevis were cultured in an ethanol concentration approximately equivalent to one drink. Techniques were developed to study various aspects of neural crest cell behaviour and a number of cellular parameters were quantified. In the presence of alcohol, a significant number of cranial neural crest cells emigrated from the explant on fibronectin but the liberation of individual cells was delayed. The cells also remained close to the explant and their morphology changed. Cranial neural crest cells did not grow on Type 1 collagen. For the purposes of comparison, the behaviour of trunk neural crest cells was also studied. The presence of alcohol correlated with increased retention of single cells on fibronectin but left other parameters unchanged. The behaviour of trunk neural crest cells growing on Type 1 collagen in the presence of alcohol did not differ from controls. Low concentrations of alcohol therefore significantly affected both cranial and trunk neural crest cells, with a wider variety of effects on cells from the cranial as opposed to the trunk region. The results suggest that low concentrations of alcohol may be more detrimental to early events in organ formation than currently suspected.
Collapse
Affiliation(s)
- Joanna Czarnobaj
- Department of Dentistry, Faculty of Medicine and Dentistry, 7020 Katz Building University of Alberta, Edmonton, Alberta, Canada T6G 2E1.
| | - Keith M Bagnall
- Department of Anatomy, Faculty of Medicine and Health Sciences, United Arab Emirates University, Box 17666 Al Ain, United Arab Emirates.
| | - J Steven Bamforth
- Department of Medical Genetics, Faculty of Medicine and Dentistry, 8-53 Medical Science Building, University of Alberta, Edmonton, Alberta, Canada T6G 2H7.
| | - Nadine C Milos
- Department of Dentistry, Faculty of Medicine and Dentistry, 7020 Katz Building University of Alberta, Edmonton, Alberta, Canada T6G 2E1.
| |
Collapse
|
40
|
EMT in developmental morphogenesis. Cancer Lett 2013; 341:9-15. [DOI: 10.1016/j.canlet.2013.02.037] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 02/14/2013] [Accepted: 02/14/2013] [Indexed: 12/24/2022]
|
41
|
Pelaez D, Huang CYC, Cheung HS. Isolation of pluripotent neural crest-derived stem cells from adult human tissues by connexin-43 enrichment. Stem Cells Dev 2013; 22:2906-14. [PMID: 23750535 DOI: 10.1089/scd.2013.0090] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Identification and isolation of pluripotent stem cells in adult tissues represent an important advancement in the fields of stem cell biology and regenerative medicine. For several years, research has been performed on the identification of biomarkers that can isolate stem cells residing in neural crest (NC)-derived adult tissues. The NC is considered a good model in stem cell biology as cells from it migrate extensively and contribute to the formation of diverse tissues in the body during organogenesis. Migration of these cells is modulated, in part, by gap junction communication among the cell sheets. Here we present a study in which, selection of connexin 43 (Cx43) expressing cells from human adult periodontal ligament yields a novel pluripotent stem cell population. Cx43⁺ periodontal ligament stem cells express pluripotency-associated transcription factors OCT4, Nanog, and Sox2, as well as NC-specific markers Sox10, p75, and Nestin. When injected in vivo into an immunodeficient mouse model, these cells were capable of generating teratomas with tissues from the three embryological germ layers: endoderm, mesoderm, and ectoderm. Furthermore, the cells formed mature structures of tissues normally arising from the NC during embryogenesis such as eccrine sweat glands of the human skin, muscle, neuronal tissues, cartilage, and bone. Immunohistochemical analysis confirmed the human origin of the neoplastic cells as well as the ectodermal and endodermal nature of some of the structures found in the tumors. These results suggest that Cx43 may be used as a biomarker to select and isolate the remnant NC pluripotent stem cells from adult human tissues arising from this embryological structure. The isolation of these cells through routine medical procedures such as wisdom teeth extraction further enhances their applicability to the regenerative medicine field.
Collapse
Affiliation(s)
- Daniel Pelaez
- 1 Geriatric Research, Education and Clinical Center , Miami VA Healthcare System, Miami, Florida
| | | | | |
Collapse
|
42
|
Hsu DM, Agarwal S, Benham A, Coarfa C, Trahan DN, Chen Z, Stowers PN, Courtney AN, Lakoma A, Barbieri E, Metelitsa LS, Gunaratne P, Kim ES, Shohet JM. G-CSF receptor positive neuroblastoma subpopulations are enriched in chemotherapy-resistant or relapsed tumors and are highly tumorigenic. Cancer Res 2013; 73:4134-46. [PMID: 23687340 DOI: 10.1158/0008-5472.can-12-4056] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Neuroblastoma is a neural crest-derived embryonal malignancy, which accounts for 13% of all pediatric cancer mortality, primarily due to tumor recurrence. Therapy-resistant cancer stem cells are implicated in tumor relapse, but definitive phenotypic evidence of the existence of these cells has been lacking. In this study, we define a highly tumorigenic subpopulation in neuroblastoma with stem cell characteristics, based on the expression of CSF3R, which encodes the receptor for granulocyte colony-stimulating factor (G-CSF). G-CSF receptor positive (aka G-CSFr(+) or CD114(+)) cells isolated from a primary tumor and the NGP cell line by flow cytometry were highly tumorigenic and capable of both self-renewal and differentiation to progeny cells. CD114(+) cells closely resembled embryonic and induced pluripotent stem cells with respect to their profiles of cell cycle, miRNA, and gene expression. In addition, they reflect a primitive undifferentiated neuroectodermal/neural crest phenotype revealing a developmental hierarchy within neuroblastoma tumors. We detected this dedifferentiated neural crest subpopulation in all established neuroblastoma cell lines, xenograft tumors, and primary tumor specimens analyzed. Ligand activation of CD114 by the addition of exogenous G-CSF to CD114(+) cells confirmed intact STAT3 upregulation, characteristic of G-CSF receptor signaling. Together, our data describe a novel distinct subpopulation within neuroblastoma with enhanced tumorigenicity and a stem cell-like phenotype, further elucidating the complex heterogeneity of solid tumors such as neuroblastoma. We propose that this subpopulation may represent an additional target for novel therapeutic approaches to this aggressive pediatric malignancy.
Collapse
Affiliation(s)
- Danielle M Hsu
- Division of Pediatric Surgery, Michael E DeBakey Department of Surgery, Section of Hematology-Oncology, Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Small cell lung cancer (SCLC) accounts for nearly 15% of human lung cancers and is one of the most aggressive solid tumors. The SCLC cells are thought to derive from self-renewing pulmonary neuroendocrine cells by oncogenic transformation. However, whether the SCLC cells possess stemness and plasticity for differentiation as normal stem cells has not been well understood thus far. In this study, we investigated the expressions of multilineage stem cell markers in the cancer cells of SCLC cell line (NCI-H446) and analyzed their clonogenicity, tumorigenicity, and plasticity for inducing differentiation. It has been found that most cancer cells of the cell line expressed multilineage stem cell markers under the routine culture conditions and generated single-cell clones in anchorage-dependent or -independent conditions. These cancer cells could form subcutaneous xenograft tumors and orthotopic lung xenograft tumors in BALB/C-nude mice. Most cells in xenograft tumors expressed stem cell markers and proliferation cell nuclear antigen Ki67, suggesting that these cancer cells remained stemness and highly proliferative ability in vivo. Intriguingly, the cancer cells could be induced to differentiate into neurons, adipocytes, and osteocytes, respectively, in vitro. During the processes of cellular phenotype-conversions, autophagy and apoptosis were two main metabolic events. There is cross-talking between autophagy and apoptosis in the differentiated cancer cells. In addition, the effects of the inhibitor and agonist for Sirtuin1/2 on the inducing osteogenic differentiation indicated that Sirtuin1/2 had an important role in this process. Taken together, these results indicate that most cancer cells of NCI-H446 cell line possess stemness and plasticity for multilineage differentiation. These findings have potentially some translational applications in treatments of SCLC with inducing differentiation therapy.
Collapse
|
44
|
Powell DR, Blasky AJ, Britt SG, Artinger KB. Riding the crest of the wave: parallels between the neural crest and cancer in epithelial-to-mesenchymal transition and migration. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2013; 5:511-22. [PMID: 23576382 PMCID: PMC3739939 DOI: 10.1002/wsbm.1224] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The neural crest (NC) is first induced as an epithelial population of cells at the neural plate border requiring complex signaling between bone morphogenetic protein, Wnt, and fibroblast growth factors to differentiate the neural and NC fate from the epidermis. Remarkably, following induction, these cells undergo an epithelial-to-mesenchymal transition (EMT), delaminate from the neural tube, and migrate through various tissue types and microenvironments before reaching their final destination where they undergo terminal differentiation. This process is mirrored in cancer metastasis, where a primary tumor will undergo an EMT before migrating and invading other cell populations to create a secondary tumor site. In recent years, as our understanding of NC EMT and migration has deepened, important new insights into tumorigenesis and metastasis have also been achieved. These discoveries have been driven by the observation that many cancers misregulate developmental genes to reacquire proliferative and migratory states. In this review, we examine how the NC provides an excellent model for studying EMT and migration. These data are discussed from the perspective of the gene regulatory networks that control both NC and cancer cell EMT and migration. Deciphering these processes in a comparative manner will expand our knowledge of the underlying etiology and pathogenesis of cancer and promote the development of novel targeted therapeutic strategies for cancer patients. © 2013 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Davalyn R Powell
- Graduate Program in Cell Biology, Stem Cells and Development, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | | | | |
Collapse
|
45
|
Strobl-Mazzulla PH, Bronner ME. A PHD12-Snail2 repressive complex epigenetically mediates neural crest epithelial-to-mesenchymal transition. ACTA ACUST UNITED AC 2013; 198:999-1010. [PMID: 22986495 PMCID: PMC3444776 DOI: 10.1083/jcb.201203098] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Neural crest cells form within the neural tube and then undergo an epithelial to mesenchymal transition (EMT) to initiate migration to distant locations. The transcriptional repressor Snail2 has been implicated in neural crest EMT via an as of yet unknown mechanism. We report that the adaptor protein PHD12 is highly expressed before neural crest EMT. At cranial levels, loss of PHD12 phenocopies Snail2 knockdown, preventing transcriptional shutdown of the adhesion molecule Cad6b (Cadherin6b), thereby inhibiting neural crest emigration. Although not directly binding to each other, PHD12 and Snail2 both directly interact with Sin3A in vivo, which in turn complexes with histone deacetylase (HDAC). Chromatin immunoprecipitation revealed that PHD12 is recruited to the Cad6b promoter during neural crest EMT. Consistent with this, lysines on histone 3 at the Cad6b promoter are hyperacetylated before neural crest emigration, correlating with active transcription, but deacetylated during EMT, reflecting the repressive state. Knockdown of either PHD12 or Snail2 prevents Cad6b promoter deacetylation. Collectively, the results show that PHD12 interacts directly with Sin3A/HDAC, which in turn interacts with Snail2, forming a complex at the Cad6b promoter and thus revealing the nature of the in vivo Snail repressive complex that regulates neural crest EMT.
Collapse
Affiliation(s)
- Pablo H Strobl-Mazzulla
- Biología del Desarrollo, Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús, Consejo Nacional de Investigaciones Científicas y Técnicas-Universidad Nacional de San Martín, 7130 Chascomús, Argentina
| | | |
Collapse
|
46
|
Nitzan E, Kalcheim C. Neural crest and somitic mesoderm as paradigms to investigate cell fate decisions during development. Dev Growth Differ 2012; 55:60-78. [PMID: 23043365 DOI: 10.1111/dgd.12004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 09/02/2012] [Accepted: 09/02/2012] [Indexed: 12/25/2022]
Abstract
The dorsal domains of the neural tube and somites are transient embryonic epithelia; they constitute the source of neural crest progenitors that generate the peripheral nervous system, pigment cells and ectomesenchyme, and of the dermomyotome that develops into myocytes, dermis and vascular cells, respectively. Based on the variety of derivatives produced by each type of epithelium, a classical yet still highly relevant question is whether these embryonic epithelia are composed of homogeneous multipotent progenitors or, alternatively, of subsets of fate-restricted cells. Growing evidence substantiates the notion that both the dorsal tube and the dermomyotome are heterogeneous epithelia composed of multipotent as well as fate-restricted precursors that emerge as such in a spatio-temporally regulated manner. Elucidation of the state of commitment of the precedent progenitors is of utmost significance for deciphering the mechanisms that regulate fate segregation during embryogenesis. In addition, it will contribute to understanding the nature of well documented neural crest-somite interactions shown to modulate the timing of neural crest cell emigration, their segmental migration, and myogenesis.
Collapse
Affiliation(s)
- Erez Nitzan
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, and Edmond and Lily Safra Center for Brain Sciences, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | | |
Collapse
|
47
|
The mechanics of metastasis: insights from a computational model. PLoS One 2012; 7:e44281. [PMID: 23028513 PMCID: PMC3460953 DOI: 10.1371/journal.pone.0044281] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 07/31/2012] [Indexed: 01/29/2023] Open
Abstract
Although it may seem obvious that mechanical forces are required to drive metastatic cell movements, understanding of the mechanical aspects of metastasis has lagged far behind genetic and biochemical knowledge. The goal of this study is to learn about the mechanics of metastasis using a cell-based finite element model that proved useful for advancing knowledge about the forces that drive embryonic cell and tissue movements. Metastasis, the predominant cause of cancer-related deaths, involves a series of mechanical events in which one or more cells dissociate from a primary tumour, migrate through normal tissue, traverse in and out of a multi-layer circulatory system vessel and resettle. The present work focuses on the dissemination steps, from dissociation to circulation. The model shows that certain surface tension relationships must be satisfied for cancerous cells to dissociate from a primary tumour and that these equations are analogous to those that govern dissociation of embryonic cells. For a dissociated cell to then migrate by invadopodium extension and contraction and exhibit the shapes seen in experiments, the invadopodium must generate a contraction equal to approximately twice that produced by the interfacial tension associated with surrounding cells. Intravasation through the wall of a vessel is governed by relationships akin to those in the previous two steps, while release from the vessel wall is governed by equations that involve surface and interfacial tensions. The model raises a number of potential research questions. It also identifies how specific mechanical properties and the sub-cellular structural components that give rise to them might be changed so as to thwart particular metastatic steps and thereby block the spread of cancer.
Collapse
|