1
|
Kim RT, Whited JL. Putative epithelial-mesenchymal transitions during salamander limb regeneration: Current perspectives and future investigations. Ann N Y Acad Sci 2024; 1540:89-103. [PMID: 39269330 PMCID: PMC11471381 DOI: 10.1111/nyas.15210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Previous studies have implicated epithelial-mesenchymal transition (EMT) in salamander limb regeneration. In this review, we describe putative roles for EMT during each stage of limb regeneration in axolotls and other salamanders. We hypothesize that EMT and EMT-like gene expression programs may regulate three main cellular processes during limb regeneration: (1) keratinocyte migration during wound closure; (2) transient invasion of the stump by epithelial cells undergoing EMT; and (3) use of EMT-like programs by non-epithelial blastemal progenitor cells to escape the confines of their niches. Finally, we propose nontraditional roles for EMT during limb regeneration that warrant further investigation, including alternative EMT regulators, stem cell activation, and fibrosis induced by aberrant EMT.
Collapse
Affiliation(s)
- Ryan T Kim
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Jessica L Whited
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
2
|
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
|
3
|
Mozin E, Massouridès E, Mournetas V, Lièvre C, Bourdon A, Jackson DL, Packer JS, Seong J, Trapnell C, Le Guiner C, Adjali O, Pinset C, Mack DL, Dupont JB. Dystrophin deficiency impairs cell junction formation during embryonic myogenesis from pluripotent stem cells. iScience 2024; 27:110242. [PMID: 39040067 PMCID: PMC11261405 DOI: 10.1016/j.isci.2024.110242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 05/02/2024] [Accepted: 06/07/2024] [Indexed: 07/24/2024] Open
Abstract
Mutations in the DMD gene lead to Duchenne muscular dystrophy (DMD), a severe neuromuscular disorder affecting young boys as they acquire motor functions. DMD is typically diagnosed at 2-4 years of age, but the absence of dystrophin has negative impacts on skeletal muscles before overt symptoms appear in patients, which poses a serious challenge in current standards of care. Here, we investigated the consequences of dystrophin deficiency during skeletal muscle development. We used single-cell transcriptome profiling to characterize the myogenic trajectory of human pluripotent stem cells and showed that DMD cells bifurcate to an alternative branch when they reach the somite stage. Dystrophin deficiency was linked to marked dysregulations of cell junction proteins involved in the cell state transitions characteristic of embryonic somitogenesis. Altogether, this work demonstrates that in vitro, dystrophin deficiency has deleterious effects on cell-cell communication during myogenic development, which should be considered in future therapeutic strategies for DMD.
Collapse
Affiliation(s)
- Elise Mozin
- Nantes Université, CHU Nantes, INSERM, TARGET, F-44000 Nantes, France
| | | | | | - Clémence Lièvre
- Nantes Université, CHU Nantes, INSERM, TARGET, F-44000 Nantes, France
| | - Audrey Bourdon
- Nantes Université, CHU Nantes, INSERM, TARGET, F-44000 Nantes, France
| | - Dana L. Jackson
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98105, USA
| | - Jonathan S. Packer
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98105, USA
| | - Juyoung Seong
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, South Korea
- Institute for Stem Cell and Regenerative Medicine, Department of Rehabilitation Medicine, University of Washington, Seattle, WA 98109, USA
| | - Cole Trapnell
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98105, USA
| | | | - Oumeya Adjali
- Nantes Université, CHU Nantes, INSERM, TARGET, F-44000 Nantes, France
| | - Christian Pinset
- Centre d’Etude des Cellules Souches, I-Stem, AFM, F-91100 Corbeil-Essonnes, France
| | - David L. Mack
- Institute for Stem Cell and Regenerative Medicine, Department of Rehabilitation Medicine, University of Washington, Seattle, WA 98109, USA
| | | |
Collapse
|
4
|
Miao Y, Pourquié O. Cellular and molecular control of vertebrate somitogenesis. Nat Rev Mol Cell Biol 2024; 25:517-533. [PMID: 38418851 PMCID: PMC11694818 DOI: 10.1038/s41580-024-00709-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2024] [Indexed: 03/02/2024]
Abstract
Segmentation is a fundamental feature of the vertebrate body plan. This metameric organization is first implemented by somitogenesis in the early embryo, when paired epithelial blocks called somites are rhythmically formed to flank the neural tube. Recent advances in in vitro models have offered new opportunities to elucidate the mechanisms that underlie somitogenesis. Notably, models derived from human pluripotent stem cells introduced an efficient proxy for studying this process during human development. In this Review, we summarize the current understanding of somitogenesis gained from both in vivo studies and in vitro studies. We deconstruct the spatiotemporal dynamics of somitogenesis into four distinct modules: dynamic events in the presomitic mesoderm, segmental determination, somite anteroposterior polarity patterning, and epithelial morphogenesis. We first focus on the segmentation clock, as well as signalling and metabolic gradients along the tissue, before discussing the clock and wavefront and other models that account for segmental determination. We then detail the molecular and cellular mechanisms of anteroposterior polarity patterning and somite epithelialization.
Collapse
Affiliation(s)
- Yuchuan Miao
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
| | - Olivier Pourquié
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
5
|
Mozin E, Massouridès E, Mournetas V, Lièvre C, Bourdon A, Jackson DL, Packer JS, Seong J, Trapnell C, Le Guiner C, Adjali O, Pinset C, Mack DL, Dupont JB. Dystrophin deficiency impairs cell junction formation during embryonic myogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.05.569919. [PMID: 38106055 PMCID: PMC10723310 DOI: 10.1101/2023.12.05.569919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Mutations in the DMD gene lead to Duchenne muscular dystrophy, a severe X-linked neuromuscular disorder that manifests itself as young boys acquire motor functions. DMD is typically diagnosed at 2 to 4 years of age, but the absence of dystrophin negatively impacts muscle structure and function before overt symptoms appear in patients, which poses a serious challenge in the optimization of standards of care. In this report, we investigated the early consequences of dystrophin deficiency during skeletal muscle development. We used single-cell transcriptome profiling to characterize the myogenic trajectory of human pluripotent stem cells and showed that DMD cells bifurcate to an alternative branch when they reach the somite stage. Here, dystrophin deficiency was linked to marked dysregulations of cell junction protein families involved in the cell state transitions characteristic of embryonic somitogenesis. Altogether, this work demonstrates that in vitro, dystrophin deficiency has deleterious effects on cell-cell communication during myogenic development, which should be considered in future therapeutic strategies for DMD.
Collapse
Affiliation(s)
- Elise Mozin
- Nantes Université, CHU Nantes, INSERM, TARGET, F-44000 Nantes, France
| | | | | | - Clémence Lièvre
- Nantes Université, CHU Nantes, INSERM, TARGET, F-44000 Nantes, France
| | - Audrey Bourdon
- Nantes Université, CHU Nantes, INSERM, TARGET, F-44000 Nantes, France
| | - Dana L Jackson
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98105, USA
| | - Jonathan S Packer
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98105, USA
| | - Juyoung Seong
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, South Korea
- Institute for Stem Cell and Regenerative Medicine, Department of Rehabilitation Medicine, University of Washington, Seattle, WA 98109, USA
| | - Cole Trapnell
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98105, USA
| | | | - Oumeya Adjali
- Nantes Université, CHU Nantes, INSERM, TARGET, F-44000 Nantes, France
| | - Christian Pinset
- Centre d’Etude des Cellules Souches, I-Stem, AFM, F-91100 Corbeil-Essonnes, France
| | - David L Mack
- Institute for Stem Cell and Regenerative Medicine, Department of Rehabilitation Medicine, University of Washington, Seattle, WA 98109, USA
| | | |
Collapse
|
6
|
Bouba I, A. Videla Rodriguez E, Smith VA, van den Brand H, Rodenburg TB, Visser B. A two-step Bayesian network approach to identify key SNPs associated to multiple phenotypic traits in four purebred laying hen lines. PLoS One 2024; 19:e0297533. [PMID: 38547081 PMCID: PMC10977676 DOI: 10.1371/journal.pone.0297533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/08/2024] [Indexed: 04/02/2024] Open
Abstract
When purebred laying hen chicks hatch, they remain at a rearing farm until approximately 17 weeks of age, after which they are transferred to a laying farm. Chicks or pullets are removed from the flocks during these 17 weeks if they display any rearing abnormality. The aim of this study was to investigate associations between single nucleotide polymorphisms (SNPs) and rearing success of 4 purebred White Leghorns layer lines by implementing a Bayesian network approach. Phenotypic traits and SNPs of four purebred genetic White Leghorn layer lines were available for 23,000 rearing batches obtained between 2010 and 2020. Associations between incubation traits (clutch size, embryo mortality), rearing traits (genetic line, first week mortality, rearing abnormalities, natural death, rearing success, pullet flock age, and season) and SNPs were analyzed, using a two-step Bayesian Network (BN) approach. Furthermore, the SNPs were connected to their corresponding genes, which were further explored in bioinformatics databases. BN analysis revealed a total of 28 SNPs associated with some of the traits: ten SNPs were associated with clutch size, another 10 with rearing abnormalities, a single SNP with natural death, and seven SNPs with first week mortality. Exploration via bioinformatics databases showed that one of the SNPs (ENAH) had a protein predicted network composed of 11 other proteins. The major hub of this SNP was CDC42 protein, which has a role in egg production and reproduction. The results highlight the power of BNs in knowledge discovery and how their application in complex biological systems can help getting a deeper understanding of functionality underlying genetic variation of rearing success in laying hens. Improved welfare and production might result from the identified SNPs. Selecting for these SNPs through breeding could reduce stress and increase livability during rearing.
Collapse
Affiliation(s)
- Ismalia Bouba
- Hendrix Genetics Research Technology & Services B.v, Hendrix Genetics, Boxmeer, North Brabant, The Netherlands
- Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | | | - V. Anne Smith
- School of Biology, University of St Andrews, St Andrews, Scotland, United Kingdom
| | - Henry van den Brand
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University & Research, Wageningen, Gelderland, The Netherlands
| | - T. Bas Rodenburg
- Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University & Research, Wageningen, Gelderland, The Netherlands
| | - Bram Visser
- Hendrix Genetics Research Technology & Services B.v, Hendrix Genetics, Boxmeer, North Brabant, The Netherlands
| |
Collapse
|
7
|
Saito S, Kanazawa U, Tatsumi A, Iida A, Takemoto T, Suzuki T. Functional analysis of a first hindlimb positioning enhancer via Gdf11 expression. Front Cell Dev Biol 2024; 12:1302141. [PMID: 38559809 PMCID: PMC10978735 DOI: 10.3389/fcell.2024.1302141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 02/22/2024] [Indexed: 04/04/2024] Open
Abstract
During the early development of tetrapods, including humans, the embryonic body elongates caudally once the anterior-posterior axis is established. During this process, region-specific vertebral morphogenesis occurs, with the determination of limb positioning along the anterior-posterior axis. We previously reported that Gdf11 functions as an anatomical integration system that determines the positioning of hindlimbs and sacral vertebrae where Gdf11 is expressed. However, the molecular mechanisms underlying induction of Gdf11 expression remain unclear. In this study, we searched for non-coding regions near the Gdf11 locus that were conserved across species to elucidate the regulatory mechanisms of Gdf11 expression. We identified an enhancer of the Gdf11 gene in intron 1 and named it highly conserved region (HCR). In HCR knockout mice, the expression level of endogenous Gdf11 was decreased, and the position of the sacral-hindlimb unit was shifted posteriorly. We also searched for factors upstream of Gdf11 based on the predicted transcription factor binding sites within the HCR. We found that inhibition of FGF signaling increased endogenous Gdf11 expression, suggesting that FGF signaling negatively regulates Gdf11 expression. However, FGF signaling does not regulate HCR activity. Our results suggest that there are species-specific Gdf11 enhancers other than HCR and that FGF signaling regulates Gdf11 expression independent of HCR.
Collapse
Affiliation(s)
- Seiji Saito
- Department of Animal Sciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Japan
| | - Utsugi Kanazawa
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Ayana Tatsumi
- Department of Biology, Graduate School of Science, Osaka Metropolitan University, Osaka, Japan
| | - Atsuo Iida
- Department of Animal Sciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Japan
| | - Tatsuya Takemoto
- Institute for Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Takayuki Suzuki
- Department of Biology, Graduate School of Science, Osaka Metropolitan University, Osaka, Japan
| |
Collapse
|
8
|
Akhurst RJ. From shape-shifting embryonic cells to oncology: The fascinating history of epithelial mesenchymal transition. Semin Cancer Biol 2023; 96:100-114. [PMID: 37852342 PMCID: PMC10883734 DOI: 10.1016/j.semcancer.2023.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 08/29/2023] [Accepted: 10/09/2023] [Indexed: 10/20/2023]
Abstract
Epithelial-to-mesenchymal transition or transformation (EMT) is a cell shape-changing process that is utilized repeatedly throughout embryogenesis and is critical to the attainment of a precise body plan. In the adult, EMT is observed under both normal and pathological conditions, such as during normal wounding healing, during development of certain fibrotic states and vascular anomalies, as well as in some cancers when malignant cells progress to become more aggressive, invasive, and metastatic. Epithelia derived from any of the three embryonic germ layers can undergo EMT, including those derived from mesoderm, such as endothelial cells (sometimes termed Endo-MT) and those derived from endoderm such as fetal liver stroma. At the cellular level, EMT is defined as the transformation of epithelial cells towards a mesenchymal phenotype and is marked by attenuation of expression of epithelial markers and de novo expression of mesenchymal markers. This process is induced by extracellular factors and can be reversible, resulting in mesenchymal-to-epithelial transformation (MET). It is now clear that a cell can simultaneously express properties of both epithelia and mesenchyme, and that such transitional cell-types drive tumor cell heterogeneity, an important aspect of cancer progression, development of a stem-like cell state, and drug resistance. Here we review some of the earliest studies demonstrating the existence of EMT during embryogenesis and discuss the discovery of the extracellular factors and intracellular signaling pathways that contribute to this process, with components of the TGFβ signaling superfamily playing a prominent role. We mention early controversies surrounding in vivo EMT during embryonic development and in adult diseased states, and the maturation of the field to a stage wherein targeting EMT to control disease states is an aspirational goal.
Collapse
Affiliation(s)
- Rosemary J Akhurst
- Department of Anatomy and UCSF Helen Diller Family Comprehensive Cancer Center, USA
| |
Collapse
|
9
|
Gredler ML, Zallen JA. Multicellular rosettes link mesenchymal-epithelial transition to radial intercalation in the mouse axial mesoderm. Dev Cell 2023:S1534-5807(23)00134-X. [PMID: 37080203 DOI: 10.1016/j.devcel.2023.03.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 01/25/2023] [Accepted: 03/24/2023] [Indexed: 04/22/2023]
Abstract
Mesenchymal-epithelial transitions are fundamental drivers of development and disease, but how these behaviors generate epithelial structure is not well understood. Here, we show that mesenchymal-epithelial transitions promote epithelial organization in the mouse node and notochordal plate through the assembly and radial intercalation of three-dimensional rosettes. Axial mesoderm rosettes acquire junctional and apical polarity, develop a central lumen, and dynamically expand, coalesce, and radially intercalate into the surface epithelium, converting mesenchymal-epithelial transitions into higher-order tissue structure. In mouse Par3 mutants, axial mesoderm rosettes establish central tight junction polarity but fail to form an expanded apical domain and lumen. These defects are associated with altered rosette dynamics, delayed radial intercalation, and formation of a small, fragmented surface epithelial structure. These results demonstrate that three-dimensional rosette behaviors translate mesenchymal-epithelial transitions into collective radial intercalation and epithelial formation, providing a strategy for building epithelial sheets from individual self-organizing units in the mammalian embryo.
Collapse
Affiliation(s)
- Marissa L Gredler
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Jennifer A Zallen
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA.
| |
Collapse
|
10
|
Abboud Asleh M, Zaher M, Asleh J, Jadon J, Shaulov L, Yelin R, Schultheiss TM. A morphogenetic wave in the chick embryo lateral mesoderm generates mesenchymal-epithelial transition through a 3D-rosette intermediate. Dev Cell 2023:S1534-5807(23)00133-8. [PMID: 37080204 DOI: 10.1016/j.devcel.2023.03.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 02/24/2023] [Accepted: 03/24/2023] [Indexed: 04/22/2023]
Abstract
Formation of epithelia through mesenchymal-epithelial transition (MET) is essential for embryonic development and for many physiological and pathological processes. This study investigates MET in vivo in the chick embryo lateral mesoderm, where a multilayered mesenchyme transforms into two parallel epithelial sheets that constitute the coelomic lining of the embryonic body cavity. Prior to MET initiation, mesenchymal cells exhibit non-polarized distribution of multiple polarity markers, albeit not aPKC. We identified an epithelializing wave that sweeps across the lateral mesoderm, the wavefront of which is characterized by the accumulation of basal fibronectin and a network of 3D rosettes composed of polarized, wedge-shaped cells surrounding a central focus of apical markers, now including aPKC. Initiation of the MET process is dependent on extracellular matrix-integrin signaling acting through focal adhesion kinase and talin, whereas progression through the rosette phase requires aPKC function. We present a stepwise model for MET, comprising polarization, 3D-rosette, and epithelialization stages.
Collapse
Affiliation(s)
- Manar Abboud Asleh
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Mira Zaher
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Jad Asleh
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Julian Jadon
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Lihi Shaulov
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Ronit Yelin
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Thomas M Schultheiss
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|
11
|
Miao Y, Djeffal Y, De Simone A, Zhu K, Lee JG, Lu Z, Silberfeld A, Rao J, Tarazona OA, Mongera A, Rigoni P, Diaz-Cuadros M, Song LMS, Di Talia S, Pourquié O. Reconstruction and deconstruction of human somitogenesis in vitro. Nature 2023; 614:500-508. [PMID: 36543321 PMCID: PMC10018515 DOI: 10.1038/s41586-022-05655-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
The vertebrate body displays a segmental organization that is most conspicuous in the periodic organization of the vertebral column and peripheral nerves. This metameric organization is first implemented when somites, which contain the precursors of skeletal muscles and vertebrae, are rhythmically generated from the presomitic mesoderm. Somites then become subdivided into anterior and posterior compartments that are essential for vertebral formation and segmental patterning of the peripheral nervous system1-4. How this key somitic subdivision is established remains poorly understood. Here we introduce three-dimensional culture systems of human pluripotent stem cells called somitoids and segmentoids, which recapitulate the formation of somite-like structures with anteroposterior identity. We identify a key function of the segmentation clock in converting temporal rhythmicity into the spatial regularity of anterior and posterior somitic compartments. We show that an initial 'salt and pepper' expression of the segmentation gene MESP2 in the newly formed segment is transformed into compartments of anterior and posterior identity through an active cell-sorting mechanism. Our research demonstrates that the major patterning modules that are involved in somitogenesis, including the clock and wavefront, anteroposterior polarity patterning and somite epithelialization, can be dissociated and operate independently in our in vitro systems. Together, we define a framework for the symmetry-breaking process that initiates somite polarity patterning. Our work provides a platform for decoding general principles of somitogenesis and advancing knowledge of human development.
Collapse
Affiliation(s)
- Yuchuan Miao
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Yannis Djeffal
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Kongju Zhu
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Jong Gwan Lee
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Ziqi Lu
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Andrew Silberfeld
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Jyoti Rao
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Oscar A Tarazona
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Alessandro Mongera
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Pietro Rigoni
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Margarete Diaz-Cuadros
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Laura Min Sook Song
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Stefano Di Talia
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Olivier Pourquié
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
12
|
Eibach S, Pang D. Junctional Neural Tube Defect (JNTD): A Rare and Relatively New Spinal Dysraphic Malformation. Adv Tech Stand Neurosurg 2023; 47:129-143. [PMID: 37640874 DOI: 10.1007/978-3-031-34981-2_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Junctional neurulation completes the sequential embryological processes of primary and secondary neurulation as the intermediary step linking the end of primary neurulation and the beginning of secondary neurulation. Its exact molecular process is a matter of ongoing scientific debate. Abnormality of junctional neurulation-junctional neural tube defect (JNTD)-was first described in 2017 based on a series of three patients who displayed a well-formed secondary neural tube, the conus, that is physically separated by a fair distance from its companion primary neural tube and functionally disconnected from rostral corticospinal control. Several other cases conforming to this bizarre neural tube arrangement have since appeared in the literature, reinforcing the validity of this entity. The clinical, neuroimaging, and electrophysiological features of JNTD, as well as the hypothesis of its embryogenetic mechanism, will be described in this chapter.
Collapse
Affiliation(s)
- Sebastian Eibach
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
- Paediatric Neurosurgery, Sydney Children's Hospital Randwick, Sydney, Australia
| | - Dachling Pang
- Great Ormond Street Hospital for Children, NHS Trust, London, UK
- Department of Paediatric Neurosurgery, University of California, Davis, USA
| |
Collapse
|
13
|
Engelhardt DM, Martyr CA, Niswander L. Pathogenesis of neural tube defects: The regulation and disruption of cellular processes underlying neural tube closure. WIREs Mech Dis 2022; 14:e1559. [PMID: 35504597 PMCID: PMC9605354 DOI: 10.1002/wsbm.1559] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 11/08/2022]
Abstract
Neural tube closure (NTC) is crucial for proper development of the brain and spinal cord and requires precise morphogenesis from a sheet of cells to an intact three-dimensional structure. NTC is dependent on successful regulation of hundreds of genes, a myriad of signaling pathways, concentration gradients, and is influenced by epigenetic and environmental cues. Failure of NTC is termed a neural tube defect (NTD) and is a leading class of congenital defects in the United States and worldwide. Though NTDs are all defined as incomplete closure of the neural tube, the pathogenesis of an NTD determines the type, severity, positioning, and accompanying phenotypes. In this review, we survey pathogenesis of NTDs relating to disruption of cellular processes arising from genetic mutations, altered epigenetic regulation, and environmental influences by micronutrients and maternal condition. This article is categorized under: Congenital Diseases > Genetics/Genomics/Epigenetics Neurological Diseases > Genetics/Genomics/Epigenetics Neurological Diseases > Stem Cells and Development.
Collapse
Affiliation(s)
- David M Engelhardt
- Molecular Cellular Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Cara A Martyr
- Molecular Cellular Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Lee Niswander
- Molecular Cellular Developmental Biology, University of Colorado, Boulder, Colorado, USA
| |
Collapse
|
14
|
Gomes de Almeida P, Rifes P, Martins-Jesus AP, Pinheiro GG, Andrade RP, Thorsteinsdóttir S. Cell–Fibronectin Interactions and Actomyosin Contractility Regulate the Segmentation Clock and Spatio-Temporal Somite Cleft Formation during Chick Embryo Somitogenesis. Cells 2022; 11:cells11132003. [PMID: 35805087 PMCID: PMC9266262 DOI: 10.3390/cells11132003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/15/2022] [Accepted: 06/21/2022] [Indexed: 12/19/2022] Open
Abstract
Fibronectin is essential for somite formation in the vertebrate embryo. Fibronectin matrix assembly starts as cells emerge from the primitive streak and ingress in the unsegmented presomitic mesoderm (PSM). PSM cells undergo cyclic waves of segmentation clock gene expression, followed by Notch-dependent upregulation of meso1 in the rostral PSM which induces somite cleft formation. However, the relevance of the fibronectin matrix for these molecular processes remains unknown. Here, we assessed the role of the PSM fibronectin matrix in the spatio-temporal regulation of chick embryo somitogenesis by perturbing (1) extracellular fibronectin matrix assembly, (2) integrin–fibronectin binding, (3) Rho-associated protein kinase (ROCK) activity and (4) non-muscle myosin II (NM II) function. We found that integrin–fibronectin engagement and NM II activity are required for cell polarization in the nascent somite. All treatments resulted in defective somitic clefts and significantly perturbed meso1 and segmentation clock gene expression in the PSM. Importantly, inhibition of actomyosin-mediated contractility increased the period of hairy1/hes4 oscillations from 90 to 120 min. Together, our work strongly suggests that the fibronectin–integrin–ROCK–NM II axis regulates segmentation clock dynamics and dictates the spatio-temporal localization of somitic clefts.
Collapse
Affiliation(s)
- Patrícia Gomes de Almeida
- cE3c—CHANGE, Departmento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, 1740-016 Lisboa, Portugal; (P.G.d.A.); (P.R.); (G.G.P.)
- ABC-RI, Algarve Biomedical Center Research Institute, 8005-139 Faro, Portugal; (A.P.M.-J.); (R.P.A.)
- Faculdade de Medicina e Ciências Biomédicas (FMCB), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Pedro Rifes
- cE3c—CHANGE, Departmento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, 1740-016 Lisboa, Portugal; (P.G.d.A.); (P.R.); (G.G.P.)
| | - Ana P. Martins-Jesus
- ABC-RI, Algarve Biomedical Center Research Institute, 8005-139 Faro, Portugal; (A.P.M.-J.); (R.P.A.)
- Faculdade de Medicina e Ciências Biomédicas (FMCB), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Gonçalo G. Pinheiro
- cE3c—CHANGE, Departmento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, 1740-016 Lisboa, Portugal; (P.G.d.A.); (P.R.); (G.G.P.)
- ABC-RI, Algarve Biomedical Center Research Institute, 8005-139 Faro, Portugal; (A.P.M.-J.); (R.P.A.)
- Faculdade de Medicina e Ciências Biomédicas (FMCB), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Raquel P. Andrade
- ABC-RI, Algarve Biomedical Center Research Institute, 8005-139 Faro, Portugal; (A.P.M.-J.); (R.P.A.)
- Faculdade de Medicina e Ciências Biomédicas (FMCB), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
- Champalimaud Research Program, Champalimaud Center for the Unknown, 1400-038 Lisboa, Portugal
| | - Sólveig Thorsteinsdóttir
- cE3c—CHANGE, Departmento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, 1740-016 Lisboa, Portugal; (P.G.d.A.); (P.R.); (G.G.P.)
- Correspondence:
| |
Collapse
|
15
|
Sanaki-Matsumiya M, Matsuda M, Gritti N, Nakaki F, Sharpe J, Trivedi V, Ebisuya M. Periodic formation of epithelial somites from human pluripotent stem cells. Nat Commun 2022; 13:2325. [PMID: 35484123 PMCID: PMC9050736 DOI: 10.1038/s41467-022-29967-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 04/11/2022] [Indexed: 12/19/2022] Open
Abstract
During embryonic development, epithelial cell blocks called somites are periodically formed according to the segmentation clock, becoming the foundation for the segmental pattern of the vertebral column. The process of somitogenesis has recently been recapitulated with murine and human pluripotent stem cells. However, an in vitro model for human somitogenesis coupled with the segmentation clock and epithelialization is still missing. Here, we report the generation of human somitoids, organoids that periodically form pairs of epithelial somite-like structures. Somitoids display clear oscillations of the segmentation clock that coincide with the segmentation of the presomitic mesoderm. The resulting somites show anterior-posterior and apical-basal polarities. Matrigel is essential for epithelialization but dispensable for the differentiation into somite cells. The size of somites is rather constant, irrespective of the initial cell number. The amount of WNT signaling instructs the proportion of mesodermal lineages in somitoids. Somitoids provide a novel platform to study human somitogenesis. Somitogenesis has been well characterized in model organisms, resulting in detailed description of the somite segmentation clock. Here they generate somitogenic organoids from human pluripotent stem cells that recapitulate somitogenesis, periodic segmentation, and proper polarity.
Collapse
Affiliation(s)
| | - Mitsuhiro Matsuda
- European Molecular Biology Laboratory (EMBL) Barcelona, Barcelona, Spain
| | - Nicola Gritti
- European Molecular Biology Laboratory (EMBL) Barcelona, Barcelona, Spain
| | - Fumio Nakaki
- European Molecular Biology Laboratory (EMBL) Barcelona, Barcelona, Spain
| | - James Sharpe
- European Molecular Biology Laboratory (EMBL) Barcelona, Barcelona, Spain
| | - Vikas Trivedi
- European Molecular Biology Laboratory (EMBL) Barcelona, Barcelona, Spain.,EMBL Heidelberg, Developmental Biology Unit, Heidelberg, Germany
| | - Miki Ebisuya
- European Molecular Biology Laboratory (EMBL) Barcelona, Barcelona, Spain.
| |
Collapse
|
16
|
Geng YW, Zhang Z, Jin H, Da JL, Zhang K, Wang JQ, Guo YY, Zhang B, Li Y. Mesenchymal-to-epithelial transition of osteoblasts induced by Fam20c knockout. Genes Genomics 2022; 44:155-164. [PMID: 35025083 DOI: 10.1007/s13258-021-01170-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/20/2021] [Indexed: 11/04/2022]
Abstract
BACKGROUND Fam20c is intimately related to tissue development and diseases. At present, it has been reported that Fam20c regulates the mineralization of osteoblasts, but there are few reports on other effects. OBJECTIVE To study the effect of Fam20c on osteoblasts by knocking out the Fam20c gene. METHODS Fam20c knockout osteoblasts were constructed by transfecting mouse osteoblasts with lentivirus. The proliferation, migration and mineralization of Fam20c knockout cells were detected by CCK-8, scratch test and alizarin red staining assays. The subcellular structure was observed by transmission electron microscopy. RT-PCR was used to detect the differential expression of mesenchymal-to-epithelial transition (MET)-related marker genes and core transcription factors. The differential expression of MET-related proteins was detected by immunofluorescence or Western blot. Transcriptome analysis of Fam20c knockout osteoblasts was performed, and real-time PCR was used to verify transcriptome analysis related to MET. RESULTS The proliferation ability of osteoblasts was not significantly changed after Fam20c deletion, but the migration ability and mineralization ability were significantly weakened. There were tight junctions between Fam20c knockout cells. The expression of mesenchymal cell marker genes and core transcription factors was significantly decreased, and the expression of epithelial cell marker genes was significantly increased. The expression of mesenchymal cell marker proteins was significantly decreased, and the expression of epithelial cell marker proteins was significantly increased. Multiple signalling molecules and pathways involved in MET have changed. CONCLUSIONS Knockdown of Fam20c resulted in MET. Fam20c affects the transcription of key factors in osteoblast MET.
Collapse
Affiliation(s)
- Ya-Wei Geng
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Zhen Zhang
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Han Jin
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Jun-Long Da
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Kai Zhang
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Jian-Qun Wang
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Yu-Yao Guo
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Bin Zhang
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, People's Republic of China. .,Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, Harbin, 150001, Heilongjiang, People's Republic of China. .,Heilongjiang Academy of Medical Sciences, Harbin, 150001, Heilongjiang, People's Republic of China.
| | - Ying Li
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, People's Republic of China. .,Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, Harbin, 150001, Heilongjiang, People's Republic of China.
| |
Collapse
|
17
|
Nychyk O, Galea GL, Molè M, Savery D, Greene NDE, Stanier P, Copp AJ. Vangl2-environment interaction causes severe neural tube defects, without abnormal neuroepithelial convergent extension. Dis Model Mech 2021; 15:273565. [PMID: 34842271 PMCID: PMC8807581 DOI: 10.1242/dmm.049194] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/19/2021] [Indexed: 11/20/2022] Open
Abstract
Planar cell polarity (PCP) signalling is vital for initiation of mouse neurulation, with diminished convergent extension (CE) cell movements leading to craniorachischisis, a severe neural tube defect (NTD). Some humans with NTDs also have PCP gene mutations but these are heterozygous, not homozygous as in mice. Other genetic or environmental factors may interact with partial loss of PCP function in human NTDs. We found that reduced sulfation of glycosaminoglycans interacts with heterozygosity for the Lp allele of Vangl2 (a core PCP gene), to cause craniorachischisis in cultured mouse embryos, with rescue by exogenous sulphate. We hypothesised this glycosaminoglycan-PCP interaction may regulate CE but, surprisingly, DiO labeling of the embryonic node demonstrates no abnormality of midline axial extension in sulfation-depleted Lp/+ embryos. Positive-control Lp/Lp embryos show severe CE defects. Abnormalities were detected in the size and shape of somites that flank the closing neural tube in sulfation-depleted Lp/+ embryos. We conclude that failure of closure initiation can arise by a mechanism other than faulty neuroepithelial CE, with possible involvement of matrix-mediated somite expansion, adjacent to the closing neural tube.
Collapse
Affiliation(s)
- Oleksandr Nychyk
- Developmental Biology & Cancer Research Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Gabriel L Galea
- Developmental Biology & Cancer Research Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Matteo Molè
- Developmental Biology & Cancer Research Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Dawn Savery
- Developmental Biology & Cancer Research Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Nicholas D E Greene
- Developmental Biology & Cancer Research Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Philip Stanier
- Genetics & Genomic Medicine Research Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Andrew J Copp
- Developmental Biology & Cancer Research Department, UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
18
|
Piatkowska AM, Evans SE, Stern CD. Cellular aspects of somite formation in vertebrates. Cells Dev 2021; 168:203732. [PMID: 34391979 DOI: 10.1016/j.cdev.2021.203732] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/04/2021] [Accepted: 08/04/2021] [Indexed: 10/20/2022]
Abstract
Vertebrate segmentation, the process that generates a regular arrangement of somites and thereby establishes the pattern of the adult body and of the musculoskeletal and peripheral nervous systems, was noticed many centuries ago. In the last few decades, there has been renewed interest in the process and especially in the molecular mechanisms that might account for its regularity and other spatial-temporal properties. Several models have been proposed but surprisingly, most of these do not provide clear links between the molecular mechanisms and the cell behaviours that generate the segmental pattern. Here we present a short survey of our current knowledge about the cellular aspects of vertebrate segmentation and the similarities and differences between different vertebrate groups in how they achieve their metameric pattern. Taking these variations into account should help to assess each of the models more appropriately.
Collapse
Affiliation(s)
- Agnieszka M Piatkowska
- Department of Cell and Developmental Biology, University College London, Gower Street (Anatomy Building), London WC1E 6BT, UK
| | - Susan E Evans
- Department of Cell and Developmental Biology, University College London, Gower Street (Anatomy Building), London WC1E 6BT, UK
| | - Claudio D Stern
- Department of Cell and Developmental Biology, University College London, Gower Street (Anatomy Building), London WC1E 6BT, UK.
| |
Collapse
|
19
|
Lachat C, Peixoto P, Hervouet E. Epithelial to Mesenchymal Transition History: From Embryonic Development to Cancers. Biomolecules 2021; 11:biom11060782. [PMID: 34067395 PMCID: PMC8224685 DOI: 10.3390/biom11060782] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/11/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a process that allows epithelial cells to progressively acquire a reversible mesenchymal phenotype. Here, we recount the main events in the history of EMT. EMT was first studied during embryonic development. Nowadays, it is an important field in cancer research, studied all around the world by more and more scientists, because it was shown that EMT is involved in cancer aggressiveness in many different ways. The main features of EMT's involvement in embryonic development, fibrosis and cancers are briefly reviewed here.
Collapse
Affiliation(s)
- Camille Lachat
- UMR 1098 RIGHT, University Bourgogne-Franche-Comté, INSERM, EFS-BFC, F-25000 Besançon, France; (P.P.); (E.H.)
- Correspondence:
| | - Paul Peixoto
- UMR 1098 RIGHT, University Bourgogne-Franche-Comté, INSERM, EFS-BFC, F-25000 Besançon, France; (P.P.); (E.H.)
- EPIgenetics and GENe EXPression Technical Platform (EPIGENExp), University Bourgogne Franche-Comté, F-25000 Besançon, France
| | - Eric Hervouet
- UMR 1098 RIGHT, University Bourgogne-Franche-Comté, INSERM, EFS-BFC, F-25000 Besançon, France; (P.P.); (E.H.)
- EPIgenetics and GENe EXPression Technical Platform (EPIGENExp), University Bourgogne Franche-Comté, F-25000 Besançon, France
- DImaCell Platform, University Bourgogne Franche-Comté, F-25000 Besançon, France
| |
Collapse
|
20
|
What we can learn from embryos to understand the mesenchymal-to-epithelial transition in tumor progression. Biochem J 2021; 478:1809-1825. [PMID: 33988704 DOI: 10.1042/bcj20210083] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/06/2021] [Accepted: 04/23/2021] [Indexed: 12/15/2022]
Abstract
Epithelial plasticity involved the terminal and transitional stages that occur during epithelial-to-mesenchymal transition (EMT) and mesenchymal-to-epithelial transition (MET), both are essential at different stages of early embryonic development that have been co-opted by cancer cells to undergo tumor metastasis. These processes are regulated at multiple instances, whereas the post-transcriptional regulation of key genes mediated by microRNAs is gaining major attention as a common and conserved pathway. In this review, we focus on discussing the latest findings of the cellular and molecular basis of the less characterized process of MET during embryonic development, with special attention to the role of microRNAs. Although we take in consideration the necessity of being cautious when extrapolating the obtained evidence, we propose some commonalities between early embryonic development and cancer progression that can shed light into our current understanding of this complex event and might aid in the design of specific therapeutic approaches.
Collapse
|
21
|
Abstract
Over 50 years after its discovery in early chick embryos, the concept of epithelial-mesenchymal transition (EMT) is now widely applied to morphogenetic studies in both physiological and pathological contexts. Indeed, the EMT field has witnessed exponential growth in recent years, driven primarily by a rapid expansion of cancer-oriented EMT research. This has led to EMT-based therapeutic interventions that bear the prospect of fighting cancer, and has given developmental biologists new impetus to investigate EMT phenomena more closely and to find suitable models to address emerging EMT-related questions. Here, and in the accompanying poster, I provide a brief summary of the current status of EMT research and give an overview of EMT models that have been used in developmental studies. I also highlight dynamic epithelialization and de-epithelialization events that are involved in many developmental processes and that should be considered to provide a broader perspective of EMT. Finally, I put forward a set of criteria to separate morphogenetic phenomena that are EMT-related from those that are not.
Collapse
Affiliation(s)
- Guojun Sheng
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| |
Collapse
|
22
|
A mechanical model of early somite segmentation. iScience 2021; 24:102317. [PMID: 33889816 PMCID: PMC8050378 DOI: 10.1016/j.isci.2021.102317] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/15/2021] [Accepted: 03/12/2021] [Indexed: 11/21/2022] Open
Abstract
Somitogenesis is often described using the clock-and-wavefront (CW) model, which does not explain how molecular signaling rearranges the pre-somitic mesoderm (PSM) cells into somites. Our scanning electron microscopy analysis of chicken embryos reveals a caudally-progressing epithelialization front in the dorsal PSM that precedes somite formation. Signs of apical constriction and tissue segmentation appear in this layer 3-4 somite lengths caudal to the last-formed somite. We propose a mechanical instability model in which a steady increase of apical contractility leads to periodic failure of adhesion junctions within the dorsal PSM and positions the future inter-somite boundaries. This model produces spatially periodic segments whose size depends on the speed of the activation front of contraction (F), and the buildup rate of contractility (Λ). The Λ/F ratio determines whether this mechanism produces spatially and temporally regular or irregular segments, and whether segment size increases with the front speed. Dorsal pre-somitic mesoderm of chicken embryos epithelializes before somite formation Dorsal epithelium shows signs of apical constriction and early segmentation A mechanical instability model can reproduce sequential segmentation A single ratio describes spatial and temporal patterns of segmentation
Collapse
|
23
|
Naganathan S, Oates A. Patterning and mechanics of somite boundaries in zebrafish embryos. Semin Cell Dev Biol 2020; 107:170-178. [DOI: 10.1016/j.semcdb.2020.04.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/12/2020] [Accepted: 04/19/2020] [Indexed: 12/12/2022]
|
24
|
Wang Z, Xia F, Labib M, Ahmadi M, Chen H, Das J, Ahmed SU, Angers S, Sargent EH, Kelley SO. Nanostructured Architectures Promote the Mesenchymal-Epithelial Transition for Invasive Cells. ACS NANO 2020; 14:5324-5336. [PMID: 32369335 DOI: 10.1021/acsnano.9b07350] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Dynamic modulation of cellular phenotypes between the epithelial and mesenchymal states-the epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET)-plays an important role in cancer progression. Nanoscale topography of culture substrates is known to affect the migration and EMT of cancer cells. However, existing platforms heavily rely on simple geometries such as grooved lines or cylindrical post arrays, which may oversimplify the complex interaction between cells and nanotopography in vivo. Here, we use electrodeposition to construct finely controlled surfaces with biomimetic fractal nanostructures as a means of examining the roles of nanotopography during the EMT/MET process. We found that nanostructures in the size range of 100 to 500 nm significantly promote MET for invasive breast and prostate cancer cells. The "METed" cells acquired distinct expression of epithelial and mesenchymal markers, displayed perturbed morphologies, and exhibited diminished migration and invasion, even after the removal of a nanotopographical stimulus. The phosphorylation of GSK-3 was decreased, which further tuned the expression of Snail and modulated the EMT/MET process. Our findings suggest that invasive cancer cells respond to the geometries and dimensions of complex nanostructured architectures.
Collapse
Affiliation(s)
- Zongjie Wang
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering, University of Toronto, Toronto, M5S 3G4, Canada
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, M5S 3G9, Canada
| | - Fan Xia
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, M5S 3M2, Canada
| | - Mahmoud Labib
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, M5S 3M2, Canada
| | - Moloud Ahmadi
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, M5S 3M2, Canada
| | - Haijie Chen
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering, University of Toronto, Toronto, M5S 3G4, Canada
| | - Jagotamoy Das
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, M5S 3M2, Canada
| | - Sharif U Ahmed
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, M5S 3M2, Canada
| | - Stéphane Angers
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, M5S 3M2, Canada
| | - Edward H Sargent
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering, University of Toronto, Toronto, M5S 3G4, Canada
| | - Shana O Kelley
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, M5S 3G9, Canada
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, M5S 3M2, Canada
- Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, M5S 1A8, Canada
| |
Collapse
|
25
|
Kawachi T, Shimokita E, Kudo R, Tadokoro R, Takahashi Y. Neural-fated self-renewing cells regulated by Sox2 during secondary neurulation in chicken tail bud. Dev Biol 2020; 461:160-171. [DOI: 10.1016/j.ydbio.2020.02.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 02/07/2020] [Indexed: 12/24/2022]
|
26
|
Eibach S, Pang D. Junctional Neural Tube Defect. J Korean Neurosurg Soc 2020; 63:327-337. [PMID: 32336064 PMCID: PMC7218194 DOI: 10.3340/jkns.2020.0018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/09/2020] [Indexed: 02/07/2023] Open
Abstract
Junctional neurulation represents the most recent adjunct to the well-known sequential embryological processes of primary and secondary neurulation. While its exact molecular processes, occurring at the end of primary and the beginning of secondary neurulation, are still being actively investigated, its pathological counterpart -junctional neural tube defect (JNTD)- had been described in 2017 based on three patients whose well-formed secondary neural tube, the conus, is widely separated from its corresponding primary neural tube and functionally disconnected from corticospinal control from above. Several other cases conforming to this bizarre neural tube arrangement have since appeared in the literature, reinforcing the validity of this entity. The cardinal clinical, neuroimaging, and electrophysiological features of JNTD, and the hypothesis of its embryogenetic mechanism, form part of this review.
Collapse
Affiliation(s)
- Sebastian Eibach
- Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia.,Department of Neurosurgery, Macquarie University Hospital, Sydney, Australia.,Department of Paediatric Neurosurgery, Sydney Children's Hospital Randwick, Sydney, Australia
| | - Dachling Pang
- Department of Paediatric Neurosurgery, Great Ormond Street Hospital for Children, NHS Trust, London, UK.,Department of Paediatric Neurosurgery, University of California, Davis, CA, USA
| |
Collapse
|
27
|
Rac1 activation in human breast carcinoma as a prognostic factor associated with therapeutic resistance. Breast Cancer 2020; 27:919-928. [PMID: 32314182 DOI: 10.1007/s12282-020-01091-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 04/02/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND RAS-related C3 botulinus toxin substrate 1 (Rac1) is a molecular switch fluctuating between GDP-bound inactive form (Rac1-GDP) and GTP-bound active form (Rac1-GTP) and involved in diverse function in both normal and malignant cells such as breast carcinoma cells. Although several studies have demonstrated immunolocalization of Rac1 protein in human breast carcinoma tissues, activation status of Rac1 still remains to be elucidated. METHODS We immunolocalized active form of Rac1 (Rac1-GTP) as well as total Rac1 using antibody specific for them in 115 invasive breast carcinoma tissues and correlated with clinicopathological parameters and clinical outcomes. RESULTS Rac1-GTP was frequently immunolocalized in the cytoplasm or cell membrane of breast carcinoma cells and it was positively correlated with Ki-67 labeling index and total Rac1 while negatively correlated with progesterone receptor. On the other hand, immunohistochemical Rac1-GTP status was significantly correlated with increased risk of recurrence and breast cancer-specific mortality of breast cancer patients and multivariate analyses did demonstrate Rac1-GTP as an independent worse prognostic factor for both disease-free and breast cancer-specific survival. In addition, Rac1-GTP was still correlated with worse prognosis in the patients who had received adjuvant chemotherapy or endocrine therapy. CONCLUSION These findings suggested Rac1 activation played pivotal roles in the progression and therapeutic resistance of breast cancers and Rac1 might be an important therapeutic target for improvement of the therapy for breast cancer patients.
Collapse
|
28
|
González N, Cardama GA, Chinestrad P, Robles-Valero J, Rodríguez-Fdez S, Lorenzo-Martín LF, Bustelo XR, Lorenzano Menna P, Gomez DE. Computational and in vitro Pharmacodynamics Characterization of 1A-116 Rac1 Inhibitor: Relevance of Trp56 in Its Biological Activity. Front Cell Dev Biol 2020; 8:240. [PMID: 32351958 PMCID: PMC7174510 DOI: 10.3389/fcell.2020.00240] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/23/2020] [Indexed: 12/20/2022] Open
Abstract
In the last years, the development of new drugs in oncology has evolved notably. In particular, drug development has shifted from empirical screening of active cytotoxic compounds to molecularly targeted drugs blocking specific biologic pathways that drive cancer progression and metastasis. Using a rational design approach, our group has developed 1A-116 as a promising Rac1 inhibitor, with antitumoral and antimetastatic effects in several types of cancer. Rac1 is over activated in a wide range of tumor types and and it is one of the most studied proteins of the Rho GTPase family. Its role in actin cytoskeleton reorganization has effects on endocytosis, vesicular trafficking, cell cycle progression and cellular migration. In this context, the regulatory activity of Rac1 affects several key processes in the course of the cancer including invasion and metastasis. The purpose of this preclinical study was to focus on the mode of action of 1A-116, conducting an interdisciplinary approach with in silico bioinformatics tools and in vitro assays. Here, we demonstrate that the tryptophan 56 residue is necessary for the inhibitory effects of 1A-116 since this compound interferes with protein-protein interactions (PPI) of Rac1GTPase involving several GEF activators. 1A-116 is also able to inhibit the oncogenic Rac1P29S mutant protein, one of the oncogenic drivers found in sun-exposed melanoma. It also inhibits numerous Rac1-regulated cellular processes such as membrane ruffling and lamellipodia formation. These results deepen our knowledge of 1A-116 inhibition of Rac1 and its biological impact on cancer progression. They also represent a good example of how in silico analyses represent a valuable approach for drug development.
Collapse
Affiliation(s)
- Nazareno González
- Laboratory of Molecular Oncology, National University of Quilmes, Bernal, Argentina
| | - Georgina A Cardama
- Laboratory of Molecular Oncology, National University of Quilmes, Bernal, Argentina.,National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Patricio Chinestrad
- Molecular Pharmacology Laboratory, National University of Quilmes, Bernal, Argentina
| | - Javier Robles-Valero
- Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Sonia Rodríguez-Fdez
- Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - L Francisco Lorenzo-Martín
- Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Xosé R Bustelo
- Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Pablo Lorenzano Menna
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina.,Molecular Pharmacology Laboratory, National University of Quilmes, Bernal, Argentina
| | - Daniel E Gomez
- Laboratory of Molecular Oncology, National University of Quilmes, Bernal, Argentina.,National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| |
Collapse
|
29
|
Saito S, Kawamura K, Matsuda Y, Suzuki T. Brilliant Blue as an alternative dye to Fast Green for in ovo electroporation. Dev Growth Differ 2019; 61:402-409. [PMID: 31612477 DOI: 10.1111/dgd.12629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/02/2019] [Accepted: 09/03/2019] [Indexed: 11/30/2022]
Abstract
Chick embryo electroporation is a powerful tool for the introduction of transgenes into tissues of interest for the study of developmental biology. This method often uses Fast Green to visualize the injected area by staining the solution containing DNA green. Here, we show that Fast Green fluoresces in a red color after electroporation, suggesting that researchers need to be cautious when detecting red fluorescence. Fast Green solution did not show any fluorescence before injection into chick embryos, but fluoresced red within 3 min post-injection into chick embryos. We identified Brilliant Blue as suitable alternative dye for use as an indicator of injection sites in ovo electroporation. We found that 0.2% of Brilliant Blue was sufficient to track the area of DNA injection. In addition, this chemical did not show red fluorescence after electroporation. Our findings demonstrate that Brilliant Blue can be used for detecting red fluorescent proteins introduced into chick embryos by electroporation. Our study also shows useful examples for the application of Brilliant Blue for the precise quantification of two fluorescence intensities after EGFP and mCherry co-electroporation.
Collapse
Affiliation(s)
- Seiji Saito
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Kazuki Kawamura
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Yoichi Matsuda
- Avian Bioscience Research Center, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Takayuki Suzuki
- Avian Bioscience Research Center, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| |
Collapse
|
30
|
Epithelial-Mesenchymal Transition Promotes the Differentiation Potential of Xenopus tropicalis Immature Sertoli Cells. Stem Cells Int 2019; 2019:8387478. [PMID: 31191685 PMCID: PMC6525813 DOI: 10.1155/2019/8387478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 03/27/2019] [Indexed: 01/18/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a fundamental process in embryonic development by which sessile epithelial cells are converted into migratory mesenchymal cells. Our laboratory has been successful in the establishment of Xenopus tropicalis immature Sertoli cells (XtiSCs) with the restricted differentiation potential. The aim of this study is the determination of factors responsible for EMT activation in XtiSCs and stemness window acquisition where cells possess the broadest differentiation potential. For this purpose, we tested three potent EMT inducers—GSK-3 inhibitor (CHIR99021), FGF2, and/or TGF-β1 ligand. XtiSCs underwent full EMT after 3-day treatment with CHIR99021 and partial EMT with FGF2 but not with TGF-β1. The morphological change of CHIR-treated XtiSCs to the typical spindle-like cell shape was associated with the upregulation of mesenchymal markers and the downregulation of epithelial markers. Moreover, only CHIR-treated XtiSCs were able to differentiate into chondrocytes in vitro and cardiomyocytes in vivo. Interestingly, EMT-shifted cells could migrate towards cancer cells (HeLa) in vitro and to the injury site in vivo. The results provide a better understanding of signaling pathways underlying the generation of testis-derived stem cells.
Collapse
|
31
|
Kho M, Shi H, Nie S. Cdc42 Effector Protein 3 Interacts With Cdc42 in Regulating Xenopus Somite Segmentation. Front Physiol 2019; 10:542. [PMID: 31133876 PMCID: PMC6514426 DOI: 10.3389/fphys.2019.00542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 04/17/2019] [Indexed: 11/15/2022] Open
Abstract
Somitogenesis is a critical process during vertebrate development that establishes the segmented body plan and gives rise to the vertebra, skeletal muscles, and dermis. While segmentation clock and wave front mechanisms have been elucidated to control the size and time of somite formation, regulation of the segmentation process that physically separates somites is not understood in detail. Here, we identified a cytoskeletal player, Cdc42 effector protein 3 (Cdc42ep3, CEP3) that is required for somite segmentation in Xenopus embryos. CEP3 is specifically expressed in somite tissue during somite segmentation. Loss-of-function experiments showed that CEP3 is not required for the specification of paraxial mesoderm, nor the differentiation of muscle cells, but is required for the segmentation process. Live imaging analysis further revealed that CEP3 is required for cell shape changes and alignment during somitogenesis. When CEP3 was knocked down, somitic cells did not elongate efficiently along the mediolateral axis and failed to undertake the 90° rotation. As a result, cells remained in a continuous sheet without an apparent segmentation cleft. CEP3 likely interacts with Cdc42 during this process, and both increased and decreased Cdc42 activity led to defective somite segmentation. Segmentation defects caused by Cdc42 knockdown can be partially rescued by the overexpression of CEP3. Conversely, loss of CEP3 resulted in the maintenance of high levels of Cdc42 activity at the cell membrane, which is normally reduced during and after somite segmentation. These results suggest that there is a feedback regulation between Cdc42 and CEP3 during somite segmentation and the activity of Cdc42 needs to be fine-tuned to control the coordinated cell shape changes and movement required for somite segmentation.
Collapse
Affiliation(s)
- Mary Kho
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States
| | - Hongyu Shi
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States
| | - Shuyi Nie
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States.,Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States.,Integrated Cancer Research Center, Georgia Institute of Technology, Atlanta, GA, United States
| |
Collapse
|
32
|
Pei D, Shu X, Gassama-Diagne A, Thiery JP. Mesenchymal–epithelial transition in development and reprogramming. Nat Cell Biol 2019; 21:44-53. [DOI: 10.1038/s41556-018-0195-z] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 08/10/2018] [Indexed: 02/07/2023]
|
33
|
Vergara HM, Ramirez J, Rosing T, Nave C, Blandino R, Saw D, Saraf P, Piexoto G, Coombes C, Adams M, Domingo CR. miR-206 is required for changes in cell adhesion that drive muscle cell morphogenesis in Xenopus laevis. Dev Biol 2018; 438:94-110. [PMID: 29596841 DOI: 10.1016/j.ydbio.2018.03.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/14/2018] [Accepted: 03/22/2018] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) are highly conserved small non-coding RNA molecules that post-transcriptionally regulate gene expression in multicellular organisms. Within the set of muscle-specific miRNAs, miR-206 expression is largely restricted to skeletal muscle and is found exclusively within the bony fish lineage. Although many studies have implicated miR-206 in muscle maintenance and disease, its role in skeletal muscle development remains largely unknown. Here, we examine the role of miR-206 during Xenopus laevis somitogenesis. In Xenopus laevis, miR-206 expression coincides with the onset of somitogenesis. We show that both knockdown and over-expression of miR-206 result in abnormal somite formation affecting muscle cell rotation, attachment, and elongation. In particular, our data suggests that miR-206 regulates changes in cell adhesion that affect the ability of newly formed somites to adhere to the notochord as well as to the intersomitic boundaries. Additionally, we show that β-dystroglycan and F-actin expression levels are significantly reduced, suggesting that knockdown of miR-206 levels affects cellular mechanics necessary for cell shape changes and attachments that are required for proper muscle formation.
Collapse
Affiliation(s)
- Hernando Martínez Vergara
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Julio Ramirez
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Trista Rosing
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Ceazar Nave
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Rebecca Blandino
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Daniel Saw
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Parag Saraf
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Gabriel Piexoto
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Coohleen Coombes
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Melissa Adams
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Carmen R Domingo
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA.
| |
Collapse
|
34
|
Gouignard N, Andrieu C, Theveneau E. Neural crest delamination and migration: Looking forward to the next 150 years. Genesis 2018; 56:e23107. [PMID: 29675839 DOI: 10.1002/dvg.23107] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 12/24/2022]
Abstract
Neural crest (NC) cells were described for the first time in 1868 by Wilhelm His. Since then, this amazing population of migratory stem cells has been intensively studied. It took a century to fully unravel their incredible abilities to contribute to nearly every organ of the body. Yet, our understanding of the cell and molecular mechanisms controlling their migration is far from complete. In this review, we summarize the current knowledge on epithelial-mesenchymal transition and collective behavior of NC cells and propose further stops at which the NC train might be calling in the near future.
Collapse
Affiliation(s)
- Nadège Gouignard
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, France
| | - Cyril Andrieu
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, France
| | - Eric Theveneau
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, France
| |
Collapse
|
35
|
Lin CY, He JY, Zeng CW, Loo MR, Chang WY, Zhang PH, Tsai HJ. microRNA-206 modulates an Rtn4a/Cxcr4a/Thbs3a axis in newly forming somites to maintain and stabilize the somite boundary formation of zebrafish embryos. Open Biol 2018; 7:rsob.170009. [PMID: 28701377 PMCID: PMC5541343 DOI: 10.1098/rsob.170009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 06/12/2017] [Indexed: 12/22/2022] Open
Abstract
Although microRNA-206 (miR-206) is known to regulate proliferation and differentiation of muscle fibroblasts, the role of miR-206 in early-stage somite development is still unknown. During somitogenesis of zebrafish embryos, reticulon4a (rtn4a) is specifically repressed by miR-206. The somite boundary was defective, and actin filaments were crossing over the boundary in either miR-206-knockdown or rtn4a-overexpressed embryos. In these treated embryos, C-X-C motif chemokine receptor 4a (cxcr4a) was reduced, while thrombospondin 3a (thbs3a) was increased. The defective boundary was phenocopied in either cxcr4a-knockdown or thbs3a-overexpressed embryos. Repression of thbs3a expression by cxcr4a reduced the occurrence of the boundary defect. We demonstrated that cxcr4a is an upstream regulator of thbs3a and that defective boundary cells could not process epithelialization in the absence of intracellular accumulation of the phosphorylated focal adhesion kinase (p-FAK) in boundary cells. Therefore, in the newly forming somites, miR-206-mediated downregulation of rtn4a increases cxcr4a. This activity largely decreases thbs3a expression in the epithelial cells of the somite boundary, which causes epithelialization of boundary cells through mesenchymal-epithelial transition (MET) and eventually leads to somite boundary formation. Collectively, we suggest that miR-206 mediates a novel pathway, the Rtn4a/Cxcr4a/Thbs3a axis, that allows boundary cells to undergo MET and form somite boundaries in the newly forming somites of zebrafish embryos.
Collapse
Affiliation(s)
- Cheng-Yung Lin
- Institute of Biomedical Sciences, Mackay Medical College, No. 46, Section 3 Zhongzhen Road, Sanzhi Dist., New Taipei City 252, Taiwan, Republic of China
| | - Jun-Yu He
- Institute of Molecular and Cellular Biology, National Taiwan University, No. 1, Section 4, Roosevelt Road, Taipei 106, Taiwan, Republic of China
| | - Chih-Wei Zeng
- Institute of Molecular and Cellular Biology, National Taiwan University, No. 1, Section 4, Roosevelt Road, Taipei 106, Taiwan, Republic of China
| | - Moo-Rumg Loo
- Institute of Molecular and Cellular Biology, National Taiwan University, No. 1, Section 4, Roosevelt Road, Taipei 106, Taiwan, Republic of China
| | - Wen-Yen Chang
- Institute of Molecular and Cellular Biology, National Taiwan University, No. 1, Section 4, Roosevelt Road, Taipei 106, Taiwan, Republic of China
| | - Po-Hsiang Zhang
- Institute of Biomedical Sciences, Mackay Medical College, No. 46, Section 3 Zhongzhen Road, Sanzhi Dist., New Taipei City 252, Taiwan, Republic of China
| | - Huai-Jen Tsai
- Institute of Biomedical Sciences, Mackay Medical College, No. 46, Section 3 Zhongzhen Road, Sanzhi Dist., New Taipei City 252, Taiwan, Republic of China
| |
Collapse
|
36
|
Chiu YJ, Hour MJ, Jin YA, Lu CC, Tsai FJ, Chen TL, Ma H, Juan YN, Yang JS. Disruption of IGF‑1R signaling by a novel quinazoline derivative, HMJ‑30, inhibits invasiveness and reverses epithelial-mesenchymal transition in osteosarcoma U‑2 OS cells. Int J Oncol 2018; 52:1465-1478. [PMID: 29568964 PMCID: PMC5873869 DOI: 10.3892/ijo.2018.4325] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 03/01/2018] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma is the most common primary malignancy of the bone and is characterized by local invasion and distant metastasis. Over the past 20 years, long-term outcomes have reached a plateau even with aggressive therapy. Overexpression of insulin-like growth factor 1 receptor (IGF‑1R) is associated with tumor proliferation, invasion and migration in osteosarcoma. In the present study, our group developed a novel quinazoline derivative, 6-fluoro‑2-(3-fluorophenyl)-4-(cyanoanilino)quinazoline (HMJ‑30), in order to disrupt IGF‑1R signaling and tumor invasiveness in osteosarcoma U‑2 OS cells. Molecular modeling, immune-precipitation, western blotting and phosphorylated protein kinase sandwich ELISA assays were used to confirm this hypothesis. The results demonstrated that HMJ‑30 selectively targeted the ATP-binding site of IGF‑1R and inhibited its downstream phosphoinositide 3-kinase/protein kinase B, Ras/mitogen-activated protein kinase, and IκK/nuclear factor-κB signaling pathways in U‑2 OS cells. HMJ‑30 inhibited U‑2 OS cell invasion and migration and downregulated protein levels and activities of matrix metalloproteinase (MMP)‑2 and MMP-9. An increase in protein levels of tissue inhibitor of metalloproteinase (TIMP)‑1 and TIMP‑2 was also observed. Furthermore, HMJ‑30 caused U‑2 OS cells to aggregate and form tight clusters, and these cells were flattened, less elongated and displayed cobblestone-like shapes. There was an increase in epithelial markers and a decrease in mesenchymal markers, indicating that the cells underwent the reverse epithelial-mesenchymal transition (EMT) process. Overall, these results demonstrated the potential molecular mechanisms underlying the effects of HMJ‑30 on invasiveness and EMT in U‑2 OS cells, suggesting that this compound deserves further investigation as a potential anti-osteosarcoma drug.
Collapse
Affiliation(s)
- Yu-Jen Chiu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Taipei Veteran General Hospital, Taipei 112, Taiwan, R.O.C
| | - Mann-Jen Hour
- School of Pharmacy, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Yi-An Jin
- Department of Dermatology, Taipei Medical University Hospital, Taipei 110, Taiwan, R.O.C
| | - Chi-Cheng Lu
- Department of Pharmacy, Buddhist Tzu Chi General Hospital, Hualien 970, Taiwan, R.O.C
| | - Fuu-Jen Tsai
- Genetics Center, Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan, R.O.C
| | - Tai-Lin Chen
- Institute of Biochemistry and Molecular Biology, National Yang Ming University, Taipei 112, Taiwan, R.O.C
| | - Hsu Ma
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Taipei Veteran General Hospital, Taipei 112, Taiwan, R.O.C
| | - Yu-Ning Juan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan, R.O.C
| |
Collapse
|
37
|
Barui A, Chowdhury F, Pandit A, Datta P. Rerouting mesenchymal stem cell trajectory towards epithelial lineage by engineering cellular niche. Biomaterials 2018; 156:28-44. [DOI: 10.1016/j.biomaterials.2017.11.036] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 10/22/2017] [Accepted: 11/21/2017] [Indexed: 02/06/2023]
|
38
|
Eibach S, Moes G, Hou YJ, Zovickian J, Pang D. Unjoined primary and secondary neural tubes: junctional neural tube defect, a new form of spinal dysraphism caused by disturbance of junctional neurulation. Childs Nerv Syst 2017; 33:1633-1647. [PMID: 27796548 DOI: 10.1007/s00381-016-3288-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 10/20/2016] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Primary and secondary neurulation are the two known processes that form the central neuraxis of vertebrates. Human phenotypes of neural tube defects (NTDs) mostly fall into two corresponding categories consistent with the two types of developmental sequence: primary NTD features an open skin defect, an exposed, unclosed neural plate (hence an open neural tube defect, or ONTD), and an unformed or poorly formed secondary neural tube, and secondary NTD with no skin abnormality (hence a closed NTD) and a malformed conus caudal to a well-developed primary neural tube. METHODS AND RESULTS We encountered three cases of a previously unrecorded form of spinal dysraphism in which the primary and secondary neural tubes are individually formed but are physically separated far apart and functionally disconnected from each other. One patient was operated on, in whom both the lumbosacral spinal cord from primary neurulation and the conus from secondary neurulation are each anatomically complete and endowed with functioning segmental motor roots tested by intraoperative triggered electromyography and direct spinal cord stimulation. The remarkable feature is that the two neural tubes are unjoined except by a functionally inert, probably non-neural band. CONCLUSION The developmental error of this peculiar malformation probably occurs during the critical transition between the end of primary and the beginning of secondary neurulation, in a stage aptly called junctional neurulation. We describe the current knowledge concerning junctional neurulation and speculate on the embryogenesis of this new class of spinal dysraphism, which we call junctional neural tube defect.
Collapse
Affiliation(s)
- Sebastian Eibach
- Paediatric Neurosurgery, Regional Centre of Paediatric Neurosurgery, Kaiser Foundation Hospitals of Northern California, Oakland, CA, USA
- Paediatric Neurosurgery, Altona Children's Hospital, Hamburg, Germany
| | - Greg Moes
- Neuropathology, Regional Centre of Paediatric Neurosurgery, Kaiser Foundation Hospitals of Northern California, Oakland, CA, USA
- Adjunct Faculty of Neuropathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yong Jin Hou
- Intraoperative Neurophysiology, Regional Centre of Paediatric Neurosurgery, Kaiser Foundation Hospitals of Northern California, Oakland, CA, USA
| | - John Zovickian
- Paediatric Neurosurgery, Regional Centre of Paediatric Neurosurgery, Kaiser Foundation Hospitals of Northern California, Oakland, CA, USA
| | - Dachling Pang
- Regional Centre of Paediatric Neurosurgery, Kaiser Foundation Hospitals of Northern California, Oakland, CA, USA.
- Paediatric Neurosurgery, University of California, Davis, CA, USA.
- Great Ormond Street Hospital for Children, NHS Trust, London, UK.
- Department of Paediatric Neurosurgery, Kaiser Permanente Medical Centre, Third Floor, Suite 39, 3600 Broadway, Oakland, CA, 94611, USA.
| |
Collapse
|
39
|
Abstract
Skeletal muscle is the largest tissue in the body and loss of its function or its regenerative properties results in debilitating musculoskeletal disorders. Understanding the mechanisms that drive skeletal muscle formation will not only help to unravel the molecular basis of skeletal muscle diseases, but also provide a roadmap for recapitulating skeletal myogenesis in vitro from pluripotent stem cells (PSCs). PSCs have become an important tool for probing developmental questions, while differentiated cell types allow the development of novel therapeutic strategies. In this Review, we provide a comprehensive overview of skeletal myogenesis from the earliest premyogenic progenitor stage to terminally differentiated myofibers, and discuss how this knowledge has been applied to differentiate PSCs into muscle fibers and their progenitors in vitro.
Collapse
Affiliation(s)
- Jérome Chal
- Department of Pathology, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Harvard Stem Cell Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Olivier Pourquié
- Department of Pathology, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, Boston, MA 02115, USA .,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Harvard Stem Cell Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, 67400 Illkirch-Graffenstaden, France
| |
Collapse
|
40
|
Cardama GA, Gonzalez N, Maggio J, Menna PL, Gomez DE. Rho GTPases as therapeutic targets in cancer (Review). Int J Oncol 2017; 51:1025-1034. [PMID: 28848995 PMCID: PMC5592879 DOI: 10.3892/ijo.2017.4093] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/17/2017] [Indexed: 12/20/2022] Open
Abstract
Rho GTPases are key molecular switches controlling the transduction of external signals to cytoplasmic and nuclear effectors. In the last few years, the development of genetic and pharmacological tools has allowed a more precise definition of the specific roles of Rho GTPases in cancer. The aim of the present review is to describe the cellular functions regulated by these proteins with focus in deregulated signals present in malignant tumors. Finally, we describe the state of the art in search of different experimental therapeutic strategies with Rho GTPases as molecular targets.
Collapse
Affiliation(s)
- G A Cardama
- Laboratory of Molecular Oncology, Department of Science and Technology, Quilmes National University, Bernal B1876BXD, Buenos Aires, Argentina
| | - N Gonzalez
- Laboratory of Molecular Oncology, Department of Science and Technology, Quilmes National University, Bernal B1876BXD, Buenos Aires, Argentina
| | - J Maggio
- Laboratory of Molecular Oncology, Department of Science and Technology, Quilmes National University, Bernal B1876BXD, Buenos Aires, Argentina
| | - P Lorenzano Menna
- Laboratory of Molecular Oncology, Department of Science and Technology, Quilmes National University, Bernal B1876BXD, Buenos Aires, Argentina
| | - D E Gomez
- Laboratory of Molecular Oncology, Department of Science and Technology, Quilmes National University, Bernal B1876BXD, Buenos Aires, Argentina
| |
Collapse
|
41
|
Marei H, Malliri A. Rac1 in human diseases: The therapeutic potential of targeting Rac1 signaling regulatory mechanisms. Small GTPases 2017; 8:139-163. [PMID: 27442895 PMCID: PMC5584733 DOI: 10.1080/21541248.2016.1211398] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 07/05/2016] [Accepted: 07/05/2016] [Indexed: 12/11/2022] Open
Abstract
Abnormal Rac1 signaling is linked to a number of debilitating human diseases, including cancer, cardiovascular diseases and neurodegenerative disorders. As such, Rac1 represents an attractive therapeutic target, yet the search for effective Rac1 inhibitors is still underway. Given the adverse effects associated with Rac1 signaling perturbation, cells have evolved several mechanisms to ensure the tight regulation of Rac1 signaling. Thus, characterizing these mechanisms can provide invaluable information regarding major cellular events that lead to aberrant Rac1 signaling. Importantly, this information can be utilized to further facilitate the development of effective pharmacological modulators that can restore normal Rac1 signaling. In this review, we focus on the pathological role of Rac1 signaling, highlighting the benefits and potential drawbacks of targeting Rac1 in a clinical setting. Additionally, we provide an overview of available compounds that target key Rac1 regulatory mechanisms and discuss future therapeutic avenues arising from our understanding of these mechanisms.
Collapse
Affiliation(s)
- Hadir Marei
- Cell Signaling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Angeliki Malliri
- Cell Signaling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| |
Collapse
|
42
|
Janesick A, Tang W, Nguyen TTL, Blumberg B. RARβ2 is required for vertebrate somitogenesis. Development 2017; 144:1997-2008. [PMID: 28432217 DOI: 10.1242/dev.144345] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 04/07/2017] [Indexed: 01/02/2023]
Abstract
During vertebrate somitogenesis, retinoic acid is known to establish the position of the determination wavefront, controlling where new somites are permitted to form along the anteroposterior body axis. Less is understood about how RAR regulates somite patterning, rostral-caudal boundary setting, specialization of myotome subdivisions or the specific RAR subtype that is required for somite patterning. Characterizing the function of RARβ has been challenging due to the absence of embryonic phenotypes in murine loss-of-function studies. Using the Xenopus system, we show that RARβ2 plays a specific role in somite number and size, restriction of the presomitic mesoderm anterior border, somite chevron morphology and hypaxial myoblast migration. Rarβ2 is the RAR subtype whose expression is most upregulated in response to ligand and its localization in the trunk somites positions it at the right time and place to respond to embryonic retinoid levels during somitogenesis. RARβ2 positively regulates Tbx3 a marker of hypaxial muscle, and negatively regulates Tbx6 via Ripply2 to restrict the anterior boundaries of the presomitic mesoderm and caudal progenitor pool. These results demonstrate for the first time an early and essential role for RARβ2 in vertebrate somitogenesis.
Collapse
Affiliation(s)
- Amanda Janesick
- Department of Developmental and Cell Biology, 2011 Biological Sciences 3, University of California, Irvine, CA 92697-2300, USA
| | - Weiyi Tang
- Department of Developmental and Cell Biology, 2011 Biological Sciences 3, University of California, Irvine, CA 92697-2300, USA
| | - Tuyen T L Nguyen
- Department of Developmental and Cell Biology, 2011 Biological Sciences 3, University of California, Irvine, CA 92697-2300, USA
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, 2011 Biological Sciences 3, University of California, Irvine, CA 92697-2300, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, USA
| |
Collapse
|
43
|
Ungefroren H, Witte D, Lehnert H. The role of small GTPases of the Rho/Rac family in TGF-β-induced EMT and cell motility in cancer. Dev Dyn 2017; 247:451-461. [DOI: 10.1002/dvdy.24505] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/30/2017] [Accepted: 04/01/2017] [Indexed: 12/12/2022] Open
Affiliation(s)
- Hendrik Ungefroren
- First Department of Medicine; University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, and University of Lübeck; Lübeck Germany
- Department of General and Thoracic Surgery; UKSH, Campus Kiel; Kiel Germany
| | - David Witte
- First Department of Medicine; University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, and University of Lübeck; Lübeck Germany
| | - Hendrik Lehnert
- First Department of Medicine; University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, and University of Lübeck; Lübeck Germany
| |
Collapse
|
44
|
Melzer C, Hass R, von der Ohe J, Lehnert H, Ungefroren H. The role of TGF-β and its crosstalk with RAC1/RAC1b signaling in breast and pancreas carcinoma. Cell Commun Signal 2017; 15:19. [PMID: 28499439 PMCID: PMC5429551 DOI: 10.1186/s12964-017-0175-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 05/08/2017] [Indexed: 12/14/2022] Open
Abstract
This article focusses on the role of TGF-β and its signaling crosstalk with the RHO family GTPases RAC1 and RAC1b in the progression of breast and pancreatic carcinoma. The aggressive nature of these tumor types is mainly due to metastatic dissemination. Metastasis is facilitated by desmoplasia, a peculiar tumor microenvironment and the ability of the tumor cells to undergo epithelial-mesenchymal transition (EMT) and to adopt a motile and invasive phenotype. These processes are controlled entirely or in part by TGF-β and the small RHO GTPase RAC1 with both proteins acting as tumor promoters in late-stage cancers. Data from our and other studies point to signaling crosstalk between TGF-β and RAC1 and the related isoform, RAC1b, in pancreatic and mammary carcinoma cells. Based on the exciting observation that RAC1b functions as an endogenous inhibitor of RAC1, we propose a model on how the relative abundance or activity of RAC1 and RAC1b in the tumor cells may determine their responses to TGF-β and, ultimately, the metastatic capacity of the tumor.
Collapse
Affiliation(s)
- Catharina Melzer
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | - Ralf Hass
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | - Juliane von der Ohe
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | - Hendrik Lehnert
- Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Campus Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Hendrik Ungefroren
- Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Campus Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
- Department of General and Thoracic Surgery, UKSH, Campus Kiel, Kiel, Germany
| |
Collapse
|
45
|
Schwabe T, Li X, Gaul U. Dynamic analysis of the mesenchymal-epithelial transition of blood-brain barrier forming glia in Drosophila. Biol Open 2017; 6:232-243. [PMID: 28108476 PMCID: PMC5312092 DOI: 10.1242/bio.020669] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
During development, many epithelia are formed by a mesenchymal-epithelial transition (MET). Here, we examine the major stages and underlying mechanisms of MET during blood-brain barrier formation in Drosophila. We show that contact with the basal lamina is essential for the growth of the barrier-forming subperineurial glia (SPG). Septate junctions (SJs), which provide insulation of the paracellular space, are not required for MET, but are necessary for the establishment of polarized SPG membrane compartments. In vivo time-lapse imaging reveals that the Moody GPCR signaling pathway regulates SPG cell growth and shape, with different levels of signaling causing distinct phenotypes. Timely, well-coordinated SPG growth is essential for the uniform insertion of SJs and thus the insulating function of the barrier. To our knowledge, this is the first dynamic in vivo analysis of all stages in the formation of a secondary epithelium, and of the key role trimeric G protein signaling plays in this important morphogenetic process. Summary: This study examines the major steps and underlying mechanisms of mesenchymal-epithelial transition of the blood-brain-barrier forming glia in Drosophila, including the role of basal lamina, septate junctions and of trimeric G protein signaling.
Collapse
Affiliation(s)
- Tina Schwabe
- Department of Biochemistry, Gene Center, Center of Integrated Protein Science (CIPSM), University of Munich, Feodor-Lynen-Str. 25, Munich 81377, Germany
| | - Xiaoling Li
- Department of Biochemistry, Gene Center, Center of Integrated Protein Science (CIPSM), University of Munich, Feodor-Lynen-Str. 25, Munich 81377, Germany.,Rockefeller University, 1230 York Ave, New York, 10065-6399 NY, USA
| | - Ulrike Gaul
- Department of Biochemistry, Gene Center, Center of Integrated Protein Science (CIPSM), University of Munich, Feodor-Lynen-Str. 25, Munich 81377, Germany
| |
Collapse
|
46
|
Chal J, Guillot C, Pourquié O. PAPC couples the segmentation clock to somite morphogenesis by regulating N-cadherin-dependent adhesion. Development 2017; 144:664-676. [PMID: 28087631 DOI: 10.1242/dev.143974] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 12/19/2016] [Indexed: 01/08/2023]
Abstract
Vertebrate segmentation is characterized by the periodic formation of epithelial somites from the mesenchymal presomitic mesoderm (PSM). How the rhythmic signaling pulse delivered by the segmentation clock is translated into the periodic morphogenesis of somites remains poorly understood. Here, we focused on the role of paraxial protocadherin (PAPC/Pcdh8) in this process. We showed that in chicken and mouse embryos, PAPC expression is tightly regulated by the clock and wavefront system in the posterior PSM. We observed that PAPC exhibits a striking complementary pattern to N-cadherin (CDH2), marking the interface of the future somite boundary in the anterior PSM. Gain and loss of function of PAPC in chicken embryos disrupted somite segmentation by altering the CDH2-dependent epithelialization of PSM cells. Our data suggest that clathrin-mediated endocytosis is increased in PAPC-expressing cells, subsequently affecting CDH2 internalization in the anterior compartment of the future somite. This in turn generates a differential adhesion interface, allowing formation of the acellular fissure that defines the somite boundary. Thus, periodic expression of PAPC in the anterior PSM triggers rhythmic endocytosis of CDH2, allowing for segmental de-adhesion and individualization of somites.
Collapse
Affiliation(s)
- Jérome Chal
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA.,Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch-Graffenstaden 67400, France.,Department of Pathology, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, USA.,Harvard Stem Cell Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Charlène Guillot
- Department of Pathology, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, USA
| | - Olivier Pourquié
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA .,Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch-Graffenstaden 67400, France.,Department of Pathology, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, USA.,Harvard Stem Cell Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA.,Howard Hughes Medical Institute, Kansas City, MO 64110, USA
| |
Collapse
|
47
|
Cruz-Solbes AS, Youker K. Epithelial to Mesenchymal Transition (EMT) and Endothelial to Mesenchymal Transition (EndMT): Role and Implications in Kidney Fibrosis. Results Probl Cell Differ 2017; 60:345-372. [PMID: 28409352 DOI: 10.1007/978-3-319-51436-9_13] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Tubulointerstitial injury is one of the hallmarks of renal disease. In particular, interstitial fibrosis has a prominent role in the development and progression of kidney injury. Collagen-producing fibroblasts are responsible for the ECM deposition. However, the origin of those activated fibroblasts is not clear. This chapter will discuss in detail the concept of epithelial to mesenchymal transition (EMT) and endothelial to mesenchymal transition (EndMT) in the context of fibrosis and kidney disease. In short, EMT and EndMT involve a change in cell shape, loss of polarity and increased motility associated with increased collagen production. Thus, providing a new source of fibroblasts. However, many controversies exist regarding the existence of EMT and EndMT in kidney disease, as well as its burden and role in disease development. The aim of this chapter is to provide an overview of the concepts and profibrotic pathways and to present the evidence that has been published in favor and against EMT and EndMT.
Collapse
|
48
|
Mishra S, Tripathi R, Singh S. Crosstalk of proteins, miRNAs involved in metastatic and epithelial–mesenchymal transition pathways. FRONTIERS IN LIFE SCIENCE 2016. [DOI: 10.1080/21553769.2016.1256843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
49
|
Nishimura T, Ito S, Saito H, Hiver S, Shigetomi K, Ikenouchi J, Takeichi M. DAAM1 stabilizes epithelial junctions by restraining WAVE complex-dependent lateral membrane motility. J Cell Biol 2016; 215:559-573. [PMID: 27807130 PMCID: PMC5119936 DOI: 10.1083/jcb.201603107] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 09/13/2016] [Accepted: 10/14/2016] [Indexed: 12/31/2022] Open
Abstract
Nishimura et al. show that DAAM1, a formin family actin polymerization regulator, stabilizes epithelial cell junctions by counteracting the WAVE complex, another actin regulator. Loss of DAAM1 promotes the motility of junctional membranes and thereby enhances their invasion of neighboring environments. Epithelial junctions comprise two subdomains, the apical junctional complex (AJC) and the adjacent lateral membrane contacts (LCs), that span the majority of the junction. The AJC is lined with circumferential actin cables, whereas the LCs are associated with less-organized actin filaments whose roles are elusive. We found that DAAM1, a formin family actin regulator, accumulated at the LCs, and its depletion caused dispersion of actin filaments at these sites while hardly affecting circumferential actin cables. DAAM1 loss enhanced the motility of LC-forming membranes, leading to their invasion of neighboring cell layers, as well as disruption of polarized epithelial layers. We found that components of the WAVE complex and its downstream targets were required for the elevation of LC motility caused by DAAM1 loss. These findings suggest that the LC membranes are motile by nature because of the WAVE complex, but DAAM1-mediated actin regulation normally restrains this motility, thereby stabilizing epithelial architecture, and that DAAM1 loss evokes invasive abilities of epithelial cells.
Collapse
Affiliation(s)
- Tamako Nishimura
- RIKEN Center for Developmental Biology, Chuo-ku, Kobe 650-0047, Japan
| | - Shoko Ito
- RIKEN Center for Developmental Biology, Chuo-ku, Kobe 650-0047, Japan
| | - Hiroko Saito
- RIKEN Center for Developmental Biology, Chuo-ku, Kobe 650-0047, Japan
| | - Sylvain Hiver
- RIKEN Center for Developmental Biology, Chuo-ku, Kobe 650-0047, Japan
| | - Kenta Shigetomi
- Department of Biology, Faculty of Sciences, Kyushu University, Nishi-Ku, Fukuoka 819-0395, Japan
| | - Junichi Ikenouchi
- Department of Biology, Faculty of Sciences, Kyushu University, Nishi-Ku, Fukuoka 819-0395, Japan
| | | |
Collapse
|
50
|
Abstract
ROR-family receptor tyrosine kinases form a small subfamily of receptor tyrosine kinases (RTKs), characterized by a conserved, unique domain architecture. ROR RTKs are evolutionary conserved throughout the animal kingdom and act as alternative receptors and coreceptors of WNT ligands. The intracellular signaling cascades activated downstream of ROR receptors are diverse, including but not limited to ROR-Frizzled-mediated activation of planar cell polarity signaling, RTK-like signaling, and antagonistic regulation of WNT/β-Catenin signaling. In line with their diverse repertoire of signaling functions, ROR receptors are involved in the regulation of multiple processes in embryonic development such as development of the axial and paraxial mesoderm, the nervous system and the neural crest, the axial and appendicular skeleton, and the kidney. In humans, mutations in the ROR2 gene cause two distinct developmental syndromes, recessive Robinow syndrome (RRS; MIM 268310) and dominant brachydactyly type B1 (BDB1; MIM 113000). In Robinow syndrome patients and animal models, the development of multiple organs is affected, whereas BDB1 results only in shortening of the distal phalanges of fingers and toes, reflecting the diversity of functions and signaling activities of ROR-family RTKs. In this chapter, we give an overview on ROR receptor structure and function. We discuss their signaling functions and role in vertebrate embryonic development with a focus on those developmental processes that are affected by mutations in the ROR2 gene in human patients.
Collapse
|