Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters










Publication year range
1.
Commun Biol ; 6(1): 1121, 2023 11 04.
Article in English | MEDLINE | ID: mdl-37925530

ABSTRACT

Skeletal muscle stem cells (MuSC) are crucial for tissue homoeostasis and repair after injury. Following activation, they proliferate to generate differentiating myoblasts. A proportion of cells self-renew, re-enter the MuSC niche under the basal lamina outside the myofiber and become quiescent. Quiescent MuSC have a primary cilium, which is disassembled upon cell cycle entry. Ex vivo experiments suggest cilia are important for MuSC self-renewal, however, their requirement for muscle regeneration in vivo remains poorly understood. Talpid3 (TA3) is essential for primary cilia formation and Hedgehog (Hh) signalling. Here we use tamoxifen-inducible conditional deletion of TA3 in MuSC (iSC-KO) and show that regeneration is impaired in response to cytotoxic injury. Depletion of MuSC after regeneration suggests impaired self-renewal, also consistent with an exacerbated phenotype in TA3iSC-KO mice after repeat injury. Single cell transcriptomics of MuSC progeny isolated from myofibers identifies components of several signalling pathways, which are deregulated in absence of TA3, including Hh and Wnt. Pharmacological activation of Wnt restores muscle regeneration, while purmorphamine, an activator of the Smoothened (Smo) co-receptor in the Hh pathway, has no effect. Together, our data show that TA3 and primary cilia are important for MuSC self-renewal and pharmacological treatment can efficiently restore muscle regeneration.


Subject(s)
Cell Cycle Proteins , Cilia , Muscles , Satellite Cells, Skeletal Muscle , Stem Cells , Animals , Mice , Cells, Cultured , Cilia/genetics , Cilia/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Muscles/cytology , Satellite Cells, Skeletal Muscle/metabolism , Cell Cycle Proteins/genetics , Stem Cells/cytology
2.
Front Pharmacol ; 14: 1264032, 2023.
Article in English | MEDLINE | ID: mdl-37860118

ABSTRACT

Introduction: Hepatocellular carcinoma (HCC) is the most common type of liver cancer, which is among the most lethal tumours. Combination therapy exploits multiple drugs to target key pathways synergistically to reduce tumour growth. Isothiocyanates have been shown to possess anticancer potential and to complement the anticancer activity of other compounds. This study aimed to investigate the potential of phenethyl isothiocyanate (PEITC) to synergise with dasatinib, improving its anticancer potential in HCC. Methods: MTT, 3D spheroids and clonogenic assays were used to assess the combination anti-tumour effect in vitro, whereas a murine syngeneic model was employed to evaluate the combination efficacy in vivo. DCFDA staining was employed to evaluate the production of reactive oxygen species (ROS), while flow cytometry and Western blot assays were used to elucidate the molecular mechanism of the synergistic activiy. Results: PEITC and dasatinib combination exhibited a synergistic effect in vitro and in vivo. The combination induced DNA damage and oxidative stress through the production of ROS, which led to the formation of a premature CDK1/Cyclin B1 complex associated with induction of mitotic catastrophe. Furthermore, ROS activated oxeiptosis, a caspase-independent form of programmed cell death. Conclusion: PEITC showed to enhance dasatinib action in treating HCC with increased production of ROS that induced cell cycle arrest followed by mitotic catastrophe, and to induce oxeiptosis. These results highlight the role that ITCs may have in cancer therapy as a complement of clinically approved chemotherapeutic drugs.

3.
Pharmaceutics ; 15(10)2023 Sep 27.
Article in English | MEDLINE | ID: mdl-37896150

ABSTRACT

Cancerous cells are characterised by their ability to invade, metastasise, and induce angiogenesis. Tumour cells use various molecules that can be targeted to reverse these processes. Dasatinib, a potent Src inhibitor, has shown promising results in treating hepatocellular carcinoma (HCC) in vitro and in vivo. However, its effectiveness is limited by focal adhesion kinase (FAK) activation. Isothiocyanates, on the other hand, are phytochemicals with broad anticancer activity and FAK inhibition capabilities. This study evaluated the synergistic effect of dasatinib and phenethyl isothiocyanate (PEITC) on HCC. The combination was tested using various assays, including MTT, adhesion, scratch, Boyden chamber, chorioallantoic membrane (CAM), and yolk sac membrane (YSM) assays to evaluate the effect of the drug combination on HCC metastatic potential and angiogenesis in vitro and in vivo. The results showed that the combination inhibited the adhesion, migration, and invasion of HepG2 cells and reduced xenograft volume in the CAM assay. Additionally, the combination reduced angiogenesis in vitro, diminishing the growth of vessels in the tube formation assay. The inhibition of FAK/STAT3 signalling led to increased E-cadherin expression and reduced VEGF secretion, reducing HCC metastatic potential. Therefore, a combination of PEITC and dasatinib could be a potential therapeutic strategy for the treatment of HCC.

4.
Dev Cell ; 58(19): 1983-1995.e7, 2023 10 09.
Article in English | MEDLINE | ID: mdl-37499658

ABSTRACT

The mammalian body plan is shaped by rhythmic segmentation of mesoderm into somites, which are transient embryonic structures that form down each side of the neural tube. We have analyzed the genome-wide transcriptional and chromatin dynamics occurring within nascent somites, from early inception of somitogenesis to the latest stages of body plan establishment. We created matched gene expression and open chromatin maps for the three leading pairs of somites at six time points during mouse embryonic development. We show that the rate of somite differentiation accelerates as development progresses. We identified a conserved maturation program followed by all somites, but somites from more developed embryos concomitantly switch on differentiation programs from derivative cell lineages soon after segmentation. Integrated analysis of the somitic transcriptional and chromatin activities identified opposing regulatory modules controlling the onset of differentiation. Our results provide a powerful, high-resolution view of the molecular genetics underlying somitic development in mammals.


Subject(s)
Embryonic Development , Somites , Pregnancy , Female , Mice , Animals , Embryonic Development/genetics , Mesoderm , Cell Differentiation/genetics , Chromatin/genetics , Mammals
5.
Biochem Soc Trans ; 50(3): 1167-1177, 2022 06 30.
Article in English | MEDLINE | ID: mdl-35604124

ABSTRACT

Mapping accessible chromatin across time scales can give insights into its dynamic nature, for example during cellular differentiation and tissue or organism development. Analysis of such data can be utilised to identify functional cis-regulatory elements (CRE) and transcription factor binding sites and, when combined with transcriptomics, can reveal gene regulatory networks (GRNs) of expressed genes. Chromatin accessibility mapping is a powerful approach and can be performed using ATAC-sequencing (ATAC-seq), whereby Tn5 transposase inserts sequencing adaptors into genomic DNA to identify differentially accessible regions of chromatin in different cell populations. It requires low sample input and can be performed and analysed relatively quickly compared with other methods. The data generated from ATAC-seq, along with other genomic approaches, can help uncover chromatin packaging and potential cis-regulatory elements that may be responsible for gene expression. Here, we describe the ATAC-seq approach and give examples from mainly vertebrate embryonic development, where such datasets have identified the highly dynamic nature of chromatin, with differing landscapes between cellular precursors for different lineages.


Subject(s)
Chromatin , High-Throughput Nucleotide Sequencing , Cell Differentiation , Gene Regulatory Networks , High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, DNA/methods
6.
Semin Cell Dev Biol ; 127: 10-16, 2022 07.
Article in English | MEDLINE | ID: mdl-34690064

ABSTRACT

A critical stage in the development of all vertebrate embryos is the generation of the body plan and its subsequent patterning and regionalisation along the main anterior-posterior axis. This includes the formation of the vertebral axial skeleton. Its organisation begins during early embryonic development with the periodic formation of paired blocks of mesoderm tissue called somites. Here, we review axial patterning of somites, with a focus on studies using amniote model systems - avian and mouse. We summarise the molecular and cellular mechanisms that generate paraxial mesoderm and review how the different anatomical regions of the vertebral column acquire their specific identity and thus shape the body plan. We also discuss the generation of organoids and embryo-like structures from embryonic stem cells, which provide insights regarding axis formation and promise to be useful for disease modelling.


Subject(s)
Mesoderm , Somites , Animals , Body Patterning , Embryonic Development , Gene Expression Regulation, Developmental , Mice , Spine , Vertebrates
7.
Nat Commun ; 12(1): 1157, 2021 02 19.
Article in English | MEDLINE | ID: mdl-33608545

ABSTRACT

Somites arising from paraxial mesoderm are a hallmark of the segmented vertebrate body plan. They form sequentially during axis extension and generate musculoskeletal cell lineages. How paraxial mesoderm becomes regionalised along the axis and how this correlates with dynamic changes of chromatin accessibility and the transcriptome remains unknown. Here, we report a spatiotemporal series of ATAC-seq and RNA-seq along the chick embryonic axis. Footprint analysis shows differential coverage of binding sites for several key transcription factors, including CDX2, LEF1 and members of HOX clusters. Associating accessible chromatin with nearby expressed genes identifies cis-regulatory elements (CRE) for TCF15 and MEOX1. We determine their spatiotemporal activity and evolutionary conservation in Xenopus and human. Epigenome silencing of endogenous CREs disrupts TCF15 and MEOX1 gene expression and recapitulates phenotypic abnormalities of anterior-posterior axis extension. Our integrated approach allows dissection of paraxial mesoderm regulatory circuits in vivo and has implications for investigating gene regulatory networks.


Subject(s)
Chick Embryo/physiology , Chromatin , Gene Expression Regulation, Developmental , Mesoderm/physiology , Regulatory Sequences, Nucleic Acid/physiology , Transcriptome , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , CDX2 Transcription Factor/genetics , CDX2 Transcription Factor/metabolism , Cell Lineage , Female , Gastrulation/genetics , Gastrulation/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Somites/metabolism , Transcription Factors/metabolism , Xenopus laevis
8.
Gene Expr Patterns ; 39: 119167, 2021 03.
Article in English | MEDLINE | ID: mdl-33460819

ABSTRACT

Frizzleds (Fzds) are transmembrane receptors that can transduce signals dependent upon binding of Wnts, a large family of secreted glycoproteins homologous to the Drosophila wingless gene. FZDs are critical for a wide variety of normal and pathological developmental processes. In the nervous system, Wnts and Frizzleds play an important role in anterior-posterior patterning, cell fate decisions, proliferation, and synaptogenesis. Here, we preformed a comprehensive expression profile of Wnt receptors (FZD) by using situ hybridization to identify FZDs that are expressed in dorsal-ventral regions of the neural tube development. Our data show specific expression for FZD1,2,3,7,9 and 10 in the chick developing spinal cord. This expression profile of cFZD receptors offers the basis for functional studies in the future to determine roles for the different FZD receptors and their interactions with Wnts during dorsal-ventral neural tube development in vivo. Furthermore, we also show that co-overexpression of Wnt1/3a by in vivo electroporation affects FZD7/10 expression in the neural tube. This illustrates an example of Wnts-FZDs interactions during spinal cord neurogenesis.


Subject(s)
Avian Proteins/genetics , Frizzled Receptors/genetics , Spinal Cord/metabolism , Animals , Avian Proteins/metabolism , Chick Embryo , Frizzled Receptors/metabolism , Gene Expression Regulation, Developmental , Spinal Cord/embryology , Wnt Proteins/genetics , Wnt Proteins/metabolism
9.
Methods Mol Biol ; 2179: 173-181, 2021.
Article in English | MEDLINE | ID: mdl-32939721

ABSTRACT

Avian (chick) embryos are an established and accessible model organism making them ideal for studying developmental processes. Chick embryos can be harvested from the egg and cultured allowing real-time observations and imaging. Here, we describe ex vivo culture and preparation of somite tissue followed by time-lapse multi-photon microscopy, image capture and processing. We applied this approach to perform live imaging of somites, the paired segments in vertebrate embryos that form in a regular sequence on either side of the neural tube, posteriorly from presomitic mesoderm (psm). Somites give rise to cell lineages of the musculoskeletal system in the trunk such as skeletal muscle, cartilage and tendon, as well as endothelial cells. Until recently it was not possible to observe the cellular dynamics underlying morphological transitions in live tissue, including in somites which undergo epithelial-to-mesenchymal transitions (EMT) during their differentiation. In addition to the experimental setup, we describe the analytical tools used for image processing.


Subject(s)
Imaging, Three-Dimensional/methods , Microscopy, Fluorescence, Multiphoton/methods , Somites/cytology , Animals , Cell Differentiation , Chick Embryo , Epithelial-Mesenchymal Transition , Image Processing, Computer-Assisted/methods , Tissue Culture Techniques/methods
10.
Dev Biol ; 469: 68-79, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33080252

ABSTRACT

MicroRNAs (miRNAs), short non-coding RNAs, which act post-transcriptionally to regulate gene expression, are of widespread significance during development and disease, including muscle disease. Advances in sequencing technology and bioinformatics led to the identification of a large number of miRNAs in vertebrates and other species, however, for many of these miRNAs specific roles have not yet been determined. LNA in situ hybridisation has revealed expression patterns of somite-enriched miRNAs, here we focus on characterising the functions of miR-128. We show that antagomiR-mediated knockdown (KD) of miR-128 in developing chick somites has a negative impact on skeletal myogenesis. Computational analysis identified the transcription factor EYA4 as a candidate target consistent with the observation that miR-128 and EYA4 display similar expression profiles. Luciferase assays confirmed that miR-128 interacts with the EYA4 3'UTR. In vivo experiments also suggest that EYA4 is regulated by miR-128. EYA4 is a member of the PAX-SIX-EYA-DACH (PSED) network of transcription factors. Therefore, we identified additional candidate miRNA binding sites in the 3'UTR of SIX1/4, EYA1/2/3 and DACH1. Using the miRanda algorithm, we found sites for miR-128, as well as for other myogenic miRNAs, miR-1a, miR-206 and miR-133a, some of these were experimentally confirmed as functional miRNA target sites. Our results reveal that miR-128 is involved in regulating skeletal myogenesis by directly targeting EYA4 with indirect effects on other PSED members, including SIX4 and PAX3. Hence, the inhibitory effect on myogenesis observed after miR-128 knockdown was rescued by concomitant knockdown of PAX3. Moreover, we show that the PSED network of transcription factors is co-regulated by multiple muscle-enriched microRNAs.


Subject(s)
Gene Expression Regulation, Developmental , MicroRNAs/metabolism , Muscle Development/genetics , Muscle, Skeletal/embryology , Transcription Factors/genetics , 3' Untranslated Regions , Animals , Chick Embryo , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Muscle, Skeletal/metabolism , PAX3 Transcription Factor/genetics , PAX3 Transcription Factor/metabolism , Somites/embryology , Somites/metabolism , Transcription Factors/metabolism
11.
Development ; 147(24)2020 12 23.
Article in English | MEDLINE | ID: mdl-33214222

ABSTRACT

Understanding how complex organ systems are assembled from simple embryonic tissues is a major challenge. Across the animal kingdom a great diversity of visual organs are initiated by a 'master control gene' called Pax6, which is both necessary and sufficient for eye development. Yet precisely how Pax6 achieves this deeply homologous function is poorly understood. Using the chick as a model organism, we show that vertebrate Pax6 interacts with a pair of morphogen-coding genes, Tgfb2 and Fst, to form a putative Turing network, which we have computationally modelled. Computer simulations suggest that this gene network is sufficient to spontaneously polarise the developing retina, establishing the first organisational axis of the eye and prefiguring its further development. Our findings reveal how retinal self-organisation may be initiated independently of the highly ordered tissue interactions that help to assemble the eye in vivo These results help to explain how stem cell aggregates spontaneously self-organise into functional eye-cups in vitro We anticipate these findings will help to underpin retinal organoid technology, which holds much promise as a platform for disease modelling, drug development and regenerative therapies.


Subject(s)
Follistatin/genetics , PAX6 Transcription Factor/genetics , Retina/growth & development , Transforming Growth Factor beta2/genetics , Animals , Cell Differentiation/genetics , Chickens/genetics , Chickens/growth & development , Eye/growth & development , Gene Expression Regulation, Developmental/genetics , Gene Regulatory Networks/genetics
12.
PLoS One ; 15(6): e0219721, 2020.
Article in English | MEDLINE | ID: mdl-32531778

ABSTRACT

Wnt/FZD signalling activity is required for spinal cord development, including the dorsal-ventral patterning of the neural tube, where it affects proliferation and specification of neurons. Wnt ligands initiate canonical, ß -catenin-dependent, signaling by binding to Frizzled receptors. However, in many developmental contexts the cognate FZD receptor for a particular Wnt ligand remains to be identified. Here, we characterized FZD10 expression in the dorsal neural tube where it overlaps with both Wnt1 and Wnt3a, as well as markers of dorsal progenitors and interneurons. We show FZD10 expression is sensitive to Wnt1, but not Wnt3a expression, and FZD10 plays a role in neural tube patterning. Knockdown approaches show that Wnt1 induced ventral expansion of dorsal neural markes, Pax6 and Pax7, requires FZD10. In contrast, Wnt3a induced dorsalization of the neural tube is not affected by FZD10 knockdown. Gain of function experiments show that FZD10 is not sufficient on its own to mediate Wnt1 activity in vivo. Indeed excess FZD10 inhibits the dorsalizing activity of Wnt1. However, addition of the Lrp6 co-receptor dramatically enhances the Wnt1/FZD10 mediated activation of dorsal markers. This suggests that the mechanism by which Wnt1 regulates proliferation and patterning in the neural tube requires both FZD10 and Lrp6.


Subject(s)
Avian Proteins/metabolism , Frizzled Receptors/metabolism , Neurogenesis , Spinal Cord/cytology , Spinal Cord/growth & development , Wnt1 Protein/metabolism , Animals , Avian Proteins/deficiency , Avian Proteins/genetics , Cell Proliferation , Chickens , Frizzled Receptors/deficiency , Frizzled Receptors/genetics , Gene Knockdown Techniques , Neural Tube/metabolism , Signal Transduction
13.
Article in English | MEDLINE | ID: mdl-31767650

ABSTRACT

Heart development is a complex process and begins with the long-range migration of cardiac progenitor cells during gastrulation. This culminates in the formation of a simple contractile tube with multiple layers, which undergoes remodeling into a four-chambered heart. During this morphogenesis, additional cell populations become incorporated. It is important to unravel the underlying genetic and cellular mechanisms to be able to identify the embryonic origin of diseases, including congenital malformations, which impair cardiac function and may affect life expectancy or quality. Owing to the evolutionary conservation of development, observations made in nonamniote and amniote vertebrate species allow us to extrapolate to human. This review will focus on the contributions made to a better understanding of heart development through studying avian embryos-mainly the chicken but also quail embryos. We will illustrate the classic and recent approaches used in the avian system, give an overview of the important discoveries made, and summarize the early stages of cardiac development up to the establishment of the four-chambered heart.


Subject(s)
Chick Embryo , Chickens/physiology , Heart/embryology , Models, Animal , Quail/embryology , Quail/physiology , Animals , Cell Culture Techniques , Cell Differentiation , Cell Lineage , Cell Movement , Cells, Cultured , Fluorescent Dyes , Heart Defects, Congenital/embryology , Heart Ventricles/embryology , Humans , Morphogenesis , Neural Crest/embryology , Organogenesis , Pericardium/embryology , Transgenes
14.
Dev Biol ; 445(2): 163-169, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30496744

ABSTRACT

BACKGROUND: Studying microRNA networks during heart development is essential to obtain a better understanding of developmental defects and diseases associated with the heart and to identify novel opportunities for therapeutics. Here we highlight the advantages of chicken embryos as a vertebrate model for studying intermediate processes of heart development. Avians develop a four-chambered heart closely resembling human anatomy and they develop ex utero, which makes them easily accessible. Furthermore, embryos are available all year with a steady supply. RESULTS: In this report we established a novel method for the knockdown of microRNA function by microinjecting AntagomiRs into the chicken heart in ovo. Our approach enables the targeted delivery of antagomirs into a locally restricted area and is not impacted by circulation. After further embryo development the successful miRNA knockdown was confirmed. Loss of function phenotypes can be evaluated rapidly, compared to more time-consuming genetic ablation experiments. The local application avoids potential systemic effects of microRNA knockdown, therefore allowing the assessment of impacts on heart development only. The method can be adjusted for different stages of chicken embryos (HH13-HH18) as well as for knockdown or targeted overexpression of coding genes. CONCLUSION: In conclusion our method allows targeted and locally restricted delivery of Antagomirs to the heart leading to successful knockdown of microRNA function. This method enables rapid phenotypic assessment, for example by gene expression analysis of multiple cardiac genes.


Subject(s)
Antagomirs/administration & dosage , Gene Knockdown Techniques/methods , Heart/embryology , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Animals , Chick Embryo , Heart Rate , Humans , Microinjections , Models, Animal , Organogenesis/genetics
15.
Sci Rep ; 8(1): 12644, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30139994

ABSTRACT

Somites are paired embryonic segments that form in a regular sequence from unsegmented mesoderm during vertebrate development. Although transient structures they are of fundamental importance as they generate cell lineages of the musculoskeletal system in the trunk such as cartilage, tendon, bone, endothelial cells and skeletal muscle. Surprisingly, very little is known about cellular dynamics underlying the morphological transitions during somite differentiation. Here, we address this by examining cellular rearrangements and morphogenesis in differentiating somites using live multi-photon imaging of transgenic chick embryos, where all cells express a membrane-bound GFP. We specifically focussed on the dynamic cellular changes in two principle regions within the somite, the medial and lateral domains, to investigate extensive morphological transformations. Furthermore, by using quantitative analysis and cell tracking, we capture for the first time a directed movement of dermomyotomal progenitor cells towards the rostro-medial domain of the dermomyotome, where skeletal muscle formation initiates.


Subject(s)
Somites/cytology , Animals , Cell Differentiation/physiology , Chick Embryo , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental/physiology , Mesoderm/cytology , Mesoderm/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Somites/metabolism
16.
Development ; 145(12)2018 06 11.
Article in English | MEDLINE | ID: mdl-29802149

ABSTRACT

Skeletal myogenesis serves as a paradigm to investigate the molecular mechanisms underlying exquisitely regulated cell fate decisions in developing embryos. The evolutionarily conserved miR-133 family of microRNAs is expressed in the myogenic lineage, but how it acts remains incompletely understood. Here, we performed genome-wide differential transcriptomics of miR-133 knockdown (KD) embryonic somites, the source of vertebrate skeletal muscle. These analyses, performed in chick embryos, revealed extensive downregulation of Sonic hedgehog (Shh) pathway components: patched receptors, Hedgehog interacting protein and the transcriptional activator Gli1. By contrast, Gli3, a transcriptional repressor, was de-repressed and confirmed as a direct miR-133 target. Phenotypically, miR-133 KD impaired myotome formation and growth by disrupting proliferation, extracellular matrix deposition and epithelialization. Together, these observations suggest that miR-133-mediated Gli3 silencing is crucial for embryonic myogenesis. Consistent with this idea, we found that activation of Shh signalling by either purmorphamine, or KD of Gli3 by antisense morpholino, rescued the miR-133 KD phenotype. Thus, we identify a novel Shh/myogenic regulatory factor/miR-133/Gli3 axis that connects epithelial morphogenesis with myogenic fate specification.


Subject(s)
Carrier Proteins/biosynthesis , Hedgehog Proteins/metabolism , Membrane Glycoproteins/biosynthesis , MicroRNAs/genetics , Muscle Development/physiology , Muscle, Skeletal/embryology , Nerve Tissue Proteins/biosynthesis , Patched Receptors/biosynthesis , Zinc Finger Protein Gli3/biosynthesis , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Chick Embryo , Down-Regulation , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Muscle Development/genetics , Muscle, Skeletal/growth & development , Primary Cell Culture , Zinc Finger Protein GLI1/biosynthesis
17.
BMC Genomics ; 19(1): 59, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29347911

ABSTRACT

BACKGROUND: The neural crest (NC) is a class of transitory stem cell-like cells unique to vertebrate embryos. NC cells arise within the dorsal neural tube where they undergo an epithelial to mesenchymal transition in order to migrate and differentiate throughout the developing embryo. The derivative cell types give rise to multiple tissues, including the craniofacial skeleton, peripheral nervous system and skin pigment cells. Several well-studied gene regulatory networks underpin NC development, which when disrupted can lead to various neurocristopathies such as craniofrontonasal dysplasia, DiGeorge syndrome and some forms of cancer. Small RNAs, such as microRNAs (miRNAs) are non-coding RNA molecules important in post-transcriptional gene silencing and critical for cellular regulation of gene expression. RESULTS: To uncover novel small RNAs in NC development we used high definition adapters and next generation sequencing of libraries derived from ectodermal explants of Xenopus laevis embryos induced to form neural and NC tissue. Ectodermal and blastula animal pole (blastula) stage tissues were also sequenced. We show that miR-427 is highly abundant in all four tissue types though in an isoform specific manner and we define a set of 11 miRNAs that are enriched in the NC. In addition, we show miR-301a and miR-338 are highly expressed in both the NC and blastula suggesting a role for these miRNAs in maintaining the stem cell-like phenotype of NC cells. CONCLUSION: We have characterised the miRNAs expressed in Xenopus embryonic explants treated to form ectoderm, neural or NC tissue. This has identified novel tissue specific miRNAs and highlighted differential expression of miR-427 isoforms.


Subject(s)
Embryo, Nonmammalian/cytology , Gene Expression Regulation, Developmental , MicroRNAs/genetics , Neural Crest/growth & development , Xenopus laevis/embryology , Animals , Base Sequence , Blastula/cytology , Blastula/metabolism , Cells, Cultured , Embryo, Nonmammalian/metabolism , Gene Regulatory Networks , Neural Crest/metabolism , Neurogenesis , Organ Specificity , Sequence Homology , Stem Cells/cytology , Stem Cells/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis/genetics
18.
Mol Neurobiol ; 55(4): 3523-3536, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28509082

ABSTRACT

Autophagy plays a very important role in numerous physiological and pathological events. However, it still remains unclear whether Atg7-induced autophagy is involved in the regulation of neural crest cell production. In this study, we found the co-location of Atg7 and Pax7+ neural crest cells in early chick embryo development. Upregulation of Atg7 with unilateral transfection of full-length Atg7 increased Pax7+ and HNK-1+ cephalic and trunk neural crest cell numbers compared to either Control-GFP transfection or opposite neural tubes, suggesting that Atg7 over-expression in neural tubes could enhance the production of neural crest cells. BMP4 in situ hybridization and p-Smad1/5/8 immunofluorescent staining demonstrated that upregulation of Atg7 in neural tubes suppressed the BMP4/Smad signaling, which is considered to promote the delamination of neural crest cells. Interestingly, upregulation of Atg7 in neural tubes could significantly accelerate cell progression into the S phase, implying that Atg7 modulates cell cycle progression. However, ß-catenin expression was not significantly altered. Finally, we demonstrated that upregulation of the Atg7 gene could activate autophagy as did Atg8. We have also observed that similar phenotypes, such as more HNK-1+ neural crest cells in the unilateral Atg8 transfection side of neural tubes, and the transfection with full-length Atg8-GFP certainly promote the numbers of BrdU+ neural crest cells in comparison to the GFP control. Taken together, we reveal that Atg7-induced autophagy is involved in regulating the production of neural crest cells in early chick embryos through the modification of the cell cycle.


Subject(s)
Autophagy-Related Protein 7/metabolism , Autophagy , Neural Crest/cytology , Neurogenesis , Animals , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 8 Family/genetics , Autophagy-Related Protein 8 Family/metabolism , Bone Morphogenetic Protein 4/metabolism , Cell Cycle , Cell Line, Tumor , Chick Embryo , Gene Expression Regulation, Developmental , Models, Biological , Neural Crest/metabolism , Neural Crest/ultrastructure , Neural Tube/cytology , Neural Tube/embryology , Neural Tube/metabolism , Neural Tube/ultrastructure , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Signal Transduction , Smad Proteins/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism
19.
Semin Cell Dev Biol ; 72: 67-76, 2017 12.
Article in English | MEDLINE | ID: mdl-29102719

ABSTRACT

A fundamental process during both embryo development and stem cell differentiation is the control of cell lineage determination. In developing skeletal muscle, many of the diffusible signaling molecules, transcription factors and more recently non-coding RNAs that contribute to this process have been identified. This has facilitated advances in our understanding of the molecular mechanisms underlying the control of cell fate choice. Here we will review the role of non-coding RNAs, in particular microRNAs (miRNAs), in embryonic muscle development and differentiation, and in satellite cells of adult muscle, which are essential for muscle growth and regeneration. Some of these short post-transcriptional regulators of gene expression are restricted to skeletal muscle, but their expression can also be more widespread. In addition, we discuss a few examples of long non-coding RNAs, which are numerous but much less well understood.


Subject(s)
Gene Expression Regulation, Developmental , MicroRNAs/genetics , Muscle Development/genetics , Muscle, Skeletal/metabolism , Animals , Cell Differentiation/genetics , Humans , Muscle, Skeletal/embryology , Muscle, Skeletal/physiology , RNA, Long Noncoding/genetics , Regeneration/genetics , Satellite Cells, Skeletal Muscle/metabolism
20.
Sci Rep ; 7(1): 12651, 2017 10 04.
Article in English | MEDLINE | ID: mdl-28978924

ABSTRACT

Angiogenesis plays an important role in hepatocellular carcinoma (HCC), the inhibition of which is explored for cancer prevention and treatment. The dietary phytochemical sulforaphane (SFN) is known for its anti-cancer properties in vitro and in vivo; but until now, no study has focused on the role of SFN in HCC tumor angiogenesis. In the present study, in vitro cell models using a HCC cell line, HepG2, and human endothelial cells, HUVECs, as well as ex vivo and in vivo models have been used to investigate the anti-tumor and anti-angiogenic effect of SFN. The results showed that SFN decreased HUVEC cell viability, migration and tube formation, all of which are important steps in angiogenesis. More importantly, SFN markedly supressed HepG2-stimulated HUVEC migration, adhesion and tube formation; which may be due to its inhibition on STAT3/HIF-1α/VEGF signalling in HepG2 cells. In addition, SFN significantly reduced HepG2 tumor growth in a modified chick embryo chorioallantoic membrane (CAM) assay, associated with a decrease of HIF-1α and VEGF expression within tumors. Collectively, these findings provide new insights into the inhibitory effect of SFN on HCC tumor angiogenesis as well as tumor growth, and indicate that SFN has potential for the prevention and treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Liver Neoplasms/genetics , STAT3 Transcription Factor/genetics , Vascular Endothelial Growth Factor A/genetics , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Chick Embryo , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Human Umbilical Vein Endothelial Cells , Humans , Isothiocyanates/pharmacology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Signal Transduction/drug effects , Sulfoxides
SELECTION OF CITATIONS
SEARCH DETAIL
...