Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 2445, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38503728

ABSTRACT

Correlative light and electron microscopy (CLEM) is an important tool for the localisation of target molecule(s) and their spatial correlation with the ultrastructural map of subcellular features at the nanometre scale. Adoption of these advanced imaging methods has been limited in plant biology, due to challenges with plant tissue permeability, fluorescence labelling efficiency, indexing of features of interest throughout the complex 3D volume and their re-localization on micrographs of ultrathin cross-sections. Here, we demonstrate an imaging approach based on tissue processing and embedding into methacrylate resin followed by imaging of sections by both, single-molecule localization microscopy and transmission electron microscopy using consecutive CLEM and same-section CLEM correlative workflow. Importantly, we demonstrate that the use of a particular type of embedding resin is not only compatible with single-molecule localization microscopy but shows improvements in the fluorophore blinking behavior relative to the whole-mount approaches. Here, we use a commercially available Click-iT ethynyl-deoxyuridine cell proliferation kit to visualize the DNA replication sites of wild-type Arabidopsis thaliana seedlings, as well as fasciata1 and nucleolin1 plants and apply our in-section CLEM imaging workflow for the analysis of S-phase progression and nucleolar organization in mutant plants with aberrant nucleolar phenotypes.


Subject(s)
Arabidopsis , Single Molecule Imaging , Microscopy, Fluorescence/methods , Microscopy, Electron , Microscopy, Electron, Transmission , Single Molecule Imaging/methods , Electrons
2.
Front Cell Dev Biol ; 10: 810527, 2022.
Article in English | MEDLINE | ID: mdl-36133922

ABSTRACT

Leucine Rich Repeat Containing G Protein-Coupled Receptor 5 (LGR5), a Wnt pathway member, has been previously recognised as a stem cell marker in numerous epithelial tissues. In this study, we used Lgr5-EGFP-CreERT2 mice to analyse the distribution of LGR5-positive cells during craniofacial development. LGR5 expressing cells were primarily located in the mesenchyme adjacent to the craniofacial epithelial structures undergoing folding, such as the nasopharyngeal duct, lingual groove, and vomeronasal organ. To follow the fate of LGR5-positive cells, we performed lineage tracing using an inducible Cre knock-in allele in combination with Rosa26-tdTomato reporter mice. The slight expansion of LGR5-positive cells was found around the vomeronasal organ, in the nasal cavity, and around the epithelium in the lingual groove. However, most LGR5 expressing cells remained in their original location, possibly supporting their signalling function for adjacent epithelium rather than exerting their role as progenitor cells for the craniofacial structures. Moreover, Lgr5 knockout mice displayed distinct defects in LGR5-positive areas, especially in the reduction of the nasopharyngeal duct, the alteration of the palatal shelves shape, abnormal epithelial folding in the lingual groove area, and the disruption of salivary gland development. The latter defect manifested as an atypical number and localisation of the glandular ducts. The gene expression of several Wnt pathway members (Rspo1-3, Axin2) was altered in Lgr5-deficient animals. However, the difference was not found in sorted EGFP-positive cells obtained from Lgr5 +/+ and Lgr5 -/- animals. Expression profiling of LGR5-positive cells revealed the expression of several markers of mesenchymal cells, antagonists, as well as agonists, of Wnt signalling, and molecules associated with the basal membrane. Therefore, LGR5-positive cells in the craniofacial area represent a very specific population of mesenchymal cells adjacent to the epithelium undergoing folding or groove formation. Our results indicate a possible novel role of LGR5 in the regulation of morphogenetic processes during the formation of complex epithelial structures in the craniofacial areas, a role which is not related to the stem cell properties of LGR5-positive cells as was previously defined for various epithelial tissues.

3.
J Appl Biomed ; 17(4): 209-217, 2019 Dec.
Article in English | MEDLINE | ID: mdl-34907719

ABSTRACT

Embryonic stem (ES) cells are pluripotent cells widely used in cell therapy and tissue engineering. However, the broader clinical applications of ES cells are limited by their genomic instability and karyotypic abnormalities. Thus, understanding the mechanisms underlying ES cell karyotypic abnormalities is critical to optimizing their clinical use. In this study, we focused on proliferating human and mouse ES cells undergoing multipolar divisions. Specifically, we analyzed the frequency and outcomes of such divisions using a combination of time-lapse microscopy and cell tracking. This revealed that cells resulting from multipolar divisions were not only viable, but they also frequently underwent subsequent cell divisions. Our novel data also showed that in human and mouse ES cells, multipolar spindles allowed more robust escape from chromosome segregation control mechanisms than bipolar spindles. Considering the frequency of multipolar divisions in proliferating ES cells, it is conceivable that cell division errors underlie ES cell karyotypic instability.

4.
Stem Cell Reports ; 11(4): 959-972, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30197118

ABSTRACT

Centrioles account for centrosomes and cilia formation. Recently, a link between centrosomal components and human developmental disorders has been established. However, the exact mechanisms how centrosome abnormalities influence embryogenesis and cell fate are not understood. PLK4-STIL module represents a key element of centrosome duplication cycle. We analyzed consequences of inactivation of the module for early events of embryogenesis in human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs). We demonstrate that blocking of PLK4 or STIL functions leads to centrosome loss followed by both p53-dependent and -independent defects, including prolonged cell divisions, upregulation of p53, chromosome instability, and, importantly, reduction of pluripotency markers and induction of differentiation. We show that the observed loss of key stem cells properties is connected to alterations in mitotic timing and protein turnover. In sum, our data define a link between centrosome, its regulators, and the control of pluripotency and differentiation in PSCs.


Subject(s)
Cell Differentiation , Cell Self Renewal , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Proliferation , Centrosome/metabolism , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Mitosis , Protein Serine-Threonine Kinases/antagonists & inhibitors
5.
Methods Mol Biol ; 1683: 149-164, 2018.
Article in English | MEDLINE | ID: mdl-29082492

ABSTRACT

In the past decade, automated microscopy has become an important tool for the drug discovery and development process. The establishment of imaging modalities as screening tools depended on technological breakthroughs in the domain of automated microscopy and automated image analysis. These types of assays are often referred to as high content screening or high content analysis (HCS/HCA). The driving force to adopt imaging for drug development is the quantity and quality of cellular information that can be collected and the enhanced physiological relevance of cellular screening compared to biochemical screening. Most imaging in drug development is performed on fixed cells as this allows uncoupling the preparation of the cells from the acquisition of the images. Live-cell imaging is technically challenging, but is very useful for many aspects of the drug development pipeline such as kinetic studies of compound mode of action or to analyze the motion of cellular components. Most vendors of HCS microscopy systems offer the option of environmental chambers and onboard pipetting on their platforms. This reflects the wish and desire of many customers to have the ability to perform live-cell assays on their HCS automated microscopes. This book chapter summarizes the challenges and advantages of live-cell imaging in drug discovery. Examples of applications are presented and the motivation to perform these assays in kinetic mode is discussed.


Subject(s)
Drug Discovery , High-Throughput Screening Assays , Animals , Cell Culture Techniques , Cells, Cultured , Drug Discovery/methods , Humans , Image Processing, Computer-Assisted , Microscopy , Molecular Imaging/methods , Software
6.
Mol Cancer Ther ; 16(9): 1831-1842, 2017 09.
Article in English | MEDLINE | ID: mdl-28619751

ABSTRACT

Checkpoint-mediated dependency of tumor cells can be deployed to selectively kill them without substantial toxicity to normal cells. Specifically, loss of CHK1, a serine threonine kinase involved in the surveillance of the G2-M checkpoint in the presence of replication stress inflicted by DNA-damaging drugs, has been reported to dramatically influence the viability of tumor cells. CHK1's pivotal role in maintaining genomic stability offers attractive opportunity for increasing the selectivity, effectivity, and reduced toxicity of chemotherapy. Some recently identified CHK1 inhibitors entered clinical trials in combination with DNA antimetabolites. Herein, we report synthesis and profiling of MU380, a nontrivial analogue of clinically profiled compound SCH900776 possessing the highly unusual N-trifluoromethylpyrazole motif, which was envisioned not to undergo metabolic oxidative dealkylation and thereby provide greater robustness to the compound. MU380 is a selective and potent inhibitor of CHK1 which sensitizes a variety of tumor cell lines to hydroxyurea or gemcitabine up to 10 times. MU380 shows extended inhibitory effects in cells, and unlike SCH900776, does not undergo in vivo N-dealkylation to the significantly less selective metabolite. Compared with SCH900776, MU380 in combination with GEM causes higher accumulation of DNA damage in tumor cells and subsequent enhanced cell death, and is more efficacious in the A2780 xenograft mouse model. Overall, MU380 represents a novel state-of-the-art CHK1 inhibitor with high potency, selectivity, and improved metabolic robustness to oxidative N-dealkylation. Mol Cancer Ther; 16(9); 1831-42. ©2017 AACR.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Checkpoint Kinase 1/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Biomarkers , Cell Cycle/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Dealkylation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Methylation , Mice , Molecular Structure , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Xenograft Model Antitumor Assays
7.
Cancer Lett ; 384: 60-69, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27693455

ABSTRACT

A significant part of current research studies utilizes various cellular models which imply specific antibiotics-containing media as well as antibiotics used for clonal selection or promoter de/activation. With the great success of developing such tools, mitochondria, once originated from bacteria, can be effectively targeted by antibiotics. For that reason, some studies propose antibiotics-targeting of mitochondria as part of anticancer therapy. Here, we have focused on the effects of various classes of antibiotics on mitochondria in cancer and non-cancer cells and demonlow mitochondrial membrane potential, reduced ATP production, altered morphology and lowered respiration rate which altogether suggested mitochondrial dysfunction (MDF). This was in parallel with increased level of reactive oxygen species (ROS) and decreased activity of mitochondrial respiration complexes. However, both survival and repopulation capacity of cancer cells was not significantly affected by the antibiotics, perhaps due to a glycolytic shift or activated autophagy. In turn, simultaneous inhibition of autophagy and treatment with antibiotics largely reduced tumorigenic properties of cancer cells suggesting potential strategy for anticancer therapy.


Subject(s)
Adenine/analogs & derivatives , Anti-Bacterial Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Autophagy/drug effects , Breast Neoplasms/drug therapy , Mitochondria/drug effects , Mitophagy/drug effects , Adenine/pharmacology , Adenosine Triphosphate/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Electron Transport Chain Complex Proteins/metabolism , Energy Metabolism/drug effects , Female , Humans , Membrane Potential, Mitochondrial/drug effects , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Time Factors , Transfection
8.
Stem Cells Dev ; 26(5): 328-340, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27863459

ABSTRACT

HMGB1 and HMGB2 proteins have been implicated in numerous cellular processes, including proliferation, differentiation, apoptosis, and tumor growth. It is unknown whether they are involved in regulating the typical functions of pluripotent human embryonic stem cells (hESCs) and/or those of the differentiated derivatives of hESCs. Using inducible, stably transfected hESCs capable of shRNA-mediated knockdown of HMGB1 and HMGB2, we provide evidence that downregulation of HMGB1 and/or HMGB2 in undifferentiated hESCs does not affect the stemness of cells and induces only minor changes to the proliferation rate, cell-cycle profile, and apoptosis. After differentiation is induced, however, the downregulation of those proteins has important effects on proliferation, apoptosis, telomerase activity, and the efficiency of differentiation toward the neuroectodermal lineage. Furthermore, those processes are affected only when one, but not both, of the two proteins is downregulated; the knockdown of both HMGB1 and HMGB2 results in a normal phenotype. Those results advance our knowledge of regulation of hESC and human neuroectodermal cell differentiation and illustrate the distinct roles of HMGB1 and HMGB2 during early human development.


Subject(s)
Cell Differentiation , HMGB1 Protein/metabolism , HMGB2 Protein/metabolism , Histones/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Apoptosis/genetics , Cell Cycle/genetics , Cell Line , Cell Lineage/genetics , Cell Proliferation/genetics , Cell Self Renewal/genetics , Cell Shape/genetics , Down-Regulation/genetics , Humans , Neural Plate/cytology , Telomerase/metabolism , Transfection
9.
Oncotarget ; 7(36): 58065-58074, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27517150

ABSTRACT

Fanconi anemia (FA) is a rare genetic disorder associated with bone-marrow failure, genome instability and cancer predisposition. Recently, we and others have demonstrated dysfunctional mitochondria with morphological alterations in FA cells accompanied by high reactive oxygen species (ROS) levels. Mitochondrial morphology is regulated by continuous fusion and fission events and the misbalance between these two is often accompanied by autophagy. Here, we provide evidence of impaired autophagy in FA. We demonstrate that FA cells have increased number of autophagic (presumably mitophagic) events and accumulate dysfunctional mitochondria due to an impaired ability to degrade them. Moreover, mitochondrial fission accompanied by oxidative stress (OS) is a prerequisite condition for mitophagy in FA and blocking this pathway may release autophagic machinery to clear dysfunctional mitochondria.


Subject(s)
Fanconi Anemia/physiopathology , Mitochondria/pathology , Mitochondrial Dynamics , Mitophagy , Rare Diseases/physiopathology , Autophagy , Cell Line , Humans , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Mitochondria/ultrastructure , Oxidative Stress , Reactive Oxygen Species/metabolism
10.
Int J Cancer ; 137(6): 1330-40, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-25735931

ABSTRACT

Ovarian cancer is one of the most common malignancies in women and contributes greatly to cancer-related deaths. Tumor suppressor candidate 3 (TUSC3) is a putative tumor suppressor gene located at chromosomal region 8p22, which is often lost in epithelial cancers. Epigenetic silencing of TUSC3 has been associated with poor prognosis, and hypermethylation of its promoter provides an independent biomarker of overall and disease-free survival in ovarian cancer patients. TUSC3 is localized to the endoplasmic reticulum in an oligosaccharyl tranferase complex responsible for the N-glycosylation of proteins. However, the precise molecular role of TUSC3 in ovarian cancer remains unclear. In this study, we establish TUSC3 as a novel ovarian cancer tumor suppressor using a xenograft mouse model and demonstrate that loss of TUSC3 alters the molecular response to endoplasmic reticulum stress and induces hallmarks of the epithelial-to-mesenchymal transition in ovarian cancer cells. In summary, we have confirmed the tumor-suppressive function of TUSC3 and identified the possible mechanism driving TUSC3-deficient ovarian cancer cells toward a malignant phenotype.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Epithelial-Mesenchymal Transition/genetics , Membrane Proteins/genetics , Ovarian Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Animals , Cell Line, Tumor , Female , Genes, Tumor Suppressor/physiology , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, SCID
11.
J Cell Sci ; 127(Pt 23): 5079-92, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25278553

ABSTRACT

The delivery of newly synthesized soluble lysosomal hydrolases to the endosomal system is essential for lysosome function and cell homeostasis. This process relies on the proper trafficking of the mannose 6-phosphate receptors (MPRs) between the trans-Golgi network (TGN), endosomes and the plasma membrane. Many transmembrane proteins regulating diverse biological processes ranging from virus production to the development of multicellular organisms also use these pathways. To explore how cell signaling modulates MPR trafficking, we used high-throughput RNA interference (RNAi) to target the human kinome and phosphatome. Using high-content image analysis, we identified 127 kinases and phosphatases belonging to different signaling networks that regulate MPR trafficking and/or the dynamic states of the subcellular compartments encountered by the MPRs. Our analysis maps the MPR trafficking pathways based on enzymes regulating phosphatidylinositol phosphate metabolism. Furthermore, it reveals how cell signaling controls the biogenesis of post-Golgi tubular carriers destined to enter the endosomal system through a SRC-dependent pathway regulating ARF1 and RAC1 signaling and myosin II activity.


Subject(s)
Cell Membrane/enzymology , Endosomes/enzymology , High-Throughput Nucleotide Sequencing/methods , RNA Interference , Receptor, IGF Type 2/metabolism , trans-Golgi Network/enzymology , ADP-Ribosylation Factor 1/genetics , ADP-Ribosylation Factor 1/metabolism , Cluster Analysis , Gene Expression Regulation, Enzymologic , Gene Regulatory Networks , HeLa Cells , Humans , Phosphatidylinositol Phosphates/metabolism , Protein Interaction Maps , Protein Transport/genetics , Receptor, IGF Type 2/genetics , Signal Transduction , Transfection , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , src-Family Kinases/genetics , src-Family Kinases/metabolism
12.
J Biomol Screen ; 19(7): 1070-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24556389

ABSTRACT

Automated imaging screens are performed mostly on fixed and stained samples to simplify the workflow and increase throughput. Some processes, such as the movement of cells and organelles or measuring membrane integrity and potential, can be measured only in living cells. Developing such assays to screen large compound or RNAi collections is challenging in many respects. Here, we develop a live-cell high-content assay for tracking endocytic organelles in medium throughput. We evaluate the added value of measuring kinetic parameters compared with measuring static parameters solely. We screened 2000 compounds in U-2 OS cells expressing Lamp1-GFP to label late endosomes. All hits have phenotypes in both static and kinetic parameters. However, we show that the kinetic parameters enable better discrimination of the mechanisms of action. Most of the compounds cause a decrease of motility of endosomes, but we identify several compounds that increase endosomal motility. In summary, we show that kinetic data help to better discriminate phenotypes and thereby obtain more subtle phenotypic clustering.


Subject(s)
Cell Migration Assays/methods , Endosomes/metabolism , Automation , Cell Line, Tumor , Combinatorial Chemistry Techniques , Green Fluorescent Proteins/chemistry , Humans , Image Processing, Computer-Assisted , Microscopy, Fluorescence , Movement , Multivariate Analysis , Nocodazole/chemistry , Phenotype , Principal Component Analysis , RNA Interference , Reproducibility of Results
13.
Exp Cell Res ; 323(1): 131-143, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24480576

ABSTRACT

Cancer cells in poorly vascularized tumor regions need to adapt to an unfavorable metabolic microenvironment. As distance from supplying blood vessels increases, oxygen and nutrient concentrations decrease and cancer cells react by stopping cell cycle progression and becoming dormant. As cytostatic drugs mainly target proliferating cells, cancer cell dormancy is considered as a major resistance mechanism to this class of anti-cancer drugs. Therefore, substances that target cancer cells in poorly vascularized tumor regions have the potential to enhance cytostatic-based chemotherapy of solid tumors. With three-dimensional growth conditions, multicellular tumor spheroids (MCTS) reproduce several parameters of the tumor microenvironment, including oxygen and nutrient gradients as well as the development of dormant tumor regions. We here report the setup of a 3D cell culture compatible high-content screening system and the identification of nine substances from two commercially available drug libraries that specifically target cells in inner MCTS core regions, while cells in outer MCTS regions or in 2D cell culture remain unaffected. We elucidated the mode of action of the identified compounds as inhibitors of the respiratory chain and show that induction of cell death in inner MCTS core regions critically depends on extracellular glucose concentrations. Finally, combinational treatment with cytostatics showed increased induction of cell death in MCTS. The data presented here shows for the first time a high-content based screening setup on 3D tumor spheroids for the identification of substances that specifically induce cell death in inner tumor spheroid core regions. This validates the approach to use 3D cell culture screening systems to identify substances that would not be detectable by 2D based screening in otherwise similar culture conditions.


Subject(s)
Antineoplastic Agents/isolation & purification , Enzyme Inhibitors/isolation & purification , Spheroids, Cellular/drug effects , Antineoplastic Agents/pharmacology , Cell Culture Techniques , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor/methods , Electron Transport/drug effects , Enzyme Inhibitors/pharmacology , Female , Glucose/metabolism , Humans , Staurosporine/pharmacology , Tumor Cells, Cultured , Tumor Microenvironment/physiology
14.
Mol Biol Cell ; 20(3): 924-36, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19056682

ABSTRACT

Lrp5/6 are crucial coreceptors for Wnt/beta-catenin signaling, a pathway biochemically distinct from noncanonical Wnt signaling pathways. Here, we examined the possible participation of Lrp5/6 in noncanonical Wnt signaling. We found that Lrp6 physically interacts with Wnt5a, but that this does not lead to phosphorylation of Lrp6 or activation of the Wnt/beta-catenin pathway. Overexpression of Lrp6 blocks activation of the Wnt5a downstream target Rac1, and this effect is dependent on intact Lrp6 extracellular domains. These results suggested that the extracellular domain of Lrp6 inhibits noncanonical Wnt signaling in vitro. In vivo, Lrp6-/- mice exhibited exencephaly and a heart phenotype. Surprisingly, these defects were rescued by deletion of Wnt5a, indicating that the phenotypes resulted from noncanonical Wnt gain-of-function. Similarly, Lrp5 and Lrp6 antisense morpholino-treated Xenopus embryos exhibited convergent extension and heart phenotypes that were rescued by knockdown of noncanonical XWnt5a and XWnt11. Thus, we provide evidence that the extracellular domains of Lrp5/6 behave as physiologically relevant inhibitors of noncanonical Wnt signaling during Xenopus and mouse development in vivo.


Subject(s)
LDL-Receptor Related Proteins/chemistry , LDL-Receptor Related Proteins/metabolism , Receptors, LDL/chemistry , Receptors, LDL/metabolism , Signal Transduction , Wnt Proteins/metabolism , Xenopus Proteins/metabolism , Animals , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Embryonic Development/drug effects , Enzyme Activation/drug effects , Gene Deletion , Heart/embryology , Heart Defects, Congenital/embryology , Heart Defects, Congenital/metabolism , Heterozygote , Low Density Lipoprotein Receptor-Related Protein-5 , Low Density Lipoprotein Receptor-Related Protein-6 , Mice , Mice, Mutant Strains , Neural Tube Defects/metabolism , Oligonucleotides, Antisense/pharmacology , Phenotype , Protein Binding/drug effects , Protein Structure, Tertiary , Receptors, LDL/deficiency , Signal Transduction/drug effects , Wnt-5a Protein , Xenopus/embryology , Xenopus/metabolism , beta Catenin/metabolism , rac1 GTP-Binding Protein/metabolism
15.
Anat Embryol (Berl) ; 211 Suppl 1: 51-6, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17039375

ABSTRACT

The transcription factors Pax3 and Pax7 are important regulators of myogenic cell fate, as demonstrated by genetic manipulations in the mouse embryo. Pax3 lies genetically upstream of MyoD and has also been shown recently to directly control Myf5 transcription in derivatives of the hypaxial somite, where it also plays an important role in ensuring cell survival. Both Pax3 and Pax7 are expressed in myogenic progenitor cells derived from the central dermomyotome that make a major contribution to skeletal muscle growth. In Pax3/Pax7 double mutants, the myogenic determination genes, Myf5 and MyoD, are not activated in these cells which become incorporated into other tissues or die. This again demonstrates the dual function of Pax factors in regulating the entry of progenitor cells into the myogenic programme and in ensuring their survival. Pax3 expression marks cells in the dermomyotome that either become myogenic or downregulate Pax3 and assume another cell fate. The latter include the smooth muscle cells of the dorsal aorta that share a common clonal origin with the skeletal muscle of the myotome, thus illustrating the initial multipotency of Pax3 expressing cells.


Subject(s)
Cell Differentiation/physiology , Gene Expression Regulation, Developmental/physiology , Mesenchymal Stem Cells/physiology , Muscles/embryology , PAX7 Transcription Factor/metabolism , Paired Box Transcription Factors/metabolism , Animals , Cell Survival/physiology , Mesenchymal Stem Cells/metabolism , Mice , MyoD Protein/metabolism , Myogenic Regulatory Factor 5/metabolism , PAX3 Transcription Factor , PAX7 Transcription Factor/genetics , Paired Box Transcription Factors/genetics
16.
Development ; 133(4): 737-49, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16436625

ABSTRACT

We show that cells of the dorsal aorta, an early blood vessel, and of the myotome, the first skeletal muscle to form within the somite, derive from a common progenitor in the mouse embryo. This conclusion is based on a retrospective clonal analysis, using a nlaacZ reporter targeted to the alpha-cardiac actin gene. A rare intragenic recombination event results in a functional nlacZ sequence, giving rise to clones of beta-galactosidase-positive cells. Periendothelial and vascular smooth muscle cells of the dorsal aorta are the main cell types labelled, demonstrating that these are clonally related to the paraxial mesoderm-derived cells of skeletal muscle. Rare endothelial cells are also seen in some clones. In younger clones, arising from a recent recombination event, myotomal labelling is predominantly in the hypaxial somite, adjacent to labelled smooth muscle cells in the aorta. Analysis of Pax3(GFP/+) embryos shows that these cells are Pax3 negative but GFP positive, with fluorescent cells in the intervening region between the aorta and the somite. This is consistent with the direct migration of smooth muscle precursor cells that had expressed Pax3. These results are discussed in terms of the paraxial mesoderm contribution to the aorta and of the mesoangioblast stem cells that derive from it.


Subject(s)
Aorta/embryology , Multipotent Stem Cells/cytology , Muscle, Skeletal/embryology , Muscle, Smooth/embryology , Paired Box Transcription Factors/metabolism , Actins/metabolism , Animals , Aorta/cytology , Cell Differentiation , Cell Lineage , Genes, Reporter , Mesoderm/physiology , Mice , Mice, Transgenic , Muscle, Skeletal/cytology , Muscle, Smooth/cytology , PAX3 Transcription Factor , Somites/cytology , beta-Galactosidase/metabolism
17.
Dev Cell ; 6(5): 685-98, 2004 May.
Article in English | MEDLINE | ID: mdl-15130493

ABSTRACT

When and how cells form and pattern the myocardium is a central issue for heart morphogenesis. Many genes are differentially expressed and function in subsets of myocardial cells. However, the lineage relationships between these cells remain poorly understood. To examine this, we have adopted a retrospective approach in the mouse embryo, based on the use of the laacZ reporter gene, targeted to the alpha-cardiac actin locus. This clonal analysis demonstrates the existence of two lineages that segregate early from a common precursor. The primitive left ventricle and the presumptive outflow tract are derived exclusively from a single lineage. Unexpectedly, all other regions of the heart, including the primitive atria, are colonized by both lineages. These results are not consistent with the prespecification of the cardiac tube as a segmented structure. They are discussed in the context of different heart fields and of the evolution of the heart.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Clone Cells/metabolism , Heart/embryology , Myocardium/cytology , Actins/metabolism , Animals , Biological Evolution , Biomarkers , Body Patterning/genetics , Clone Cells/cytology , Gene Expression Regulation, Developmental/genetics , Genes, Reporter , Lac Operon , Mice , Stem Cells/cytology , Stem Cells/metabolism
18.
J Cell Biol ; 164(1): 97-109, 2004 Jan 05.
Article in English | MEDLINE | ID: mdl-14709543

ABSTRACT

During heart morphogenesis, cardiac chambers arise by differential expansion of regions of the primitive cardiac tube. This process is under the control of specific transcription factors such as Tbx5 and dHAND. To gain insight into the cellular mechanisms that underlie cardiogenesis, we have used a retrospective clonal approach based on the spontaneous recombination of an nlaacZ reporter gene targeted to the murine alpha-cardiac actin locus. We show that clonal growth of myocardial cells is oriented. At embryonic day (E) 10.5, the shape of clones is characteristic of a given cardiac region and reflects its morphology. This is already detectable in the primitive cardiac tube at E8.5, and is maintained after septation at E14.5 with additional modulations. The clonal analysis reveals new subdivisions of the myocardium, including an interventricular boundary region. Our results show that the myocardium, from the time of its formation, is a polarized and regionalized tissue and point to the role of oriented clonal cell growth in cardiac chamber morphogenesis.


Subject(s)
Cell Polarity/genetics , Clone Cells/metabolism , Heart/embryology , Myocardium/metabolism , Organogenesis/genetics , Actins/genetics , Animals , Cell Differentiation/genetics , Cell Division/genetics , Cell Size/genetics , Clone Cells/cytology , Genes, Reporter/genetics , Heart/physiology , Heart Atria/embryology , Heart Ventricles/embryology , Lac Operon/genetics , Mice , Mice, Transgenic , Myocardium/cytology , Organogenesis/physiology , Ventricular Function
19.
Mol Reprod Dev ; 65(3): 245-53, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12784245

ABSTRACT

Enzymes of the Polo-like kinase (Plk) family are active in the pathways controlling mitosis in several species. We have cloned cDNA fragments of the porcine homologues of Plk1, Plk2, and Plk3 employing fetal fibroblasts as source. All three partial cDNAs showed high sequence homology with their mouse and human counterparts and contained the Polo box, a domain characteristic for all Polo kinases. The expression levels of Plk1 mRNA at various points of the cell cycle in synchronized porcine fetal fibroblasts were analyzed by both RT-PCR and the ribonuclease protection assay. Plk1 mRNA was barely detectable in G0 and G1, increased during S phase and peaked after the G2/M transition. A monoclonal antibody was generated against an in vitro expressed porcine Plk1-protein fragment and used to detect changes in Plk1 expression at the protein level. Plk1 protein was first detected by immunoblotting at the beginning of S phase and was highest after the G2/M transition. In summary, the Plk1 expression pattern in the pig is similar to that reported for other species. The absence of Plk1 mRNA and protein appears to be a good marker for G0/G1 and thus for the selection of donor cells for nuclear transfer based somatic cloning.


Subject(s)
Embryonic and Fetal Development/physiology , Fibroblasts/cytology , Gene Expression Regulation, Developmental , Protein Kinases/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cloning, Molecular , DNA Primers , Fetus , Fibroblasts/physiology , HeLa Cells , Humans , Mice , Molecular Sequence Data , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid , Swine , Tumor Suppressor Proteins , Xenopus , Xenopus Proteins/genetics , Polo-Like Kinase 1
20.
Int J Dev Biol ; 46(6): 817-25, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12382948

ABSTRACT

The cellular response to fibroblast growth factors (FGFs) is mediated by receptor tyrosine kinases (FGFR-1 - 4) whose patterns of expression are spatially and temporally restricted during embryogenesis. These receptors have differential ligand binding capacities and are coupled to diverse signalling pathways. In the present study, we have characterized the ability of FGFR-1-deficient mouse embryonic stem (ES) cells to bind FGF-2 and to proliferate in the absence or presence of exogenous FGF-2. Under the same conditions, we also analysed the differentiation of FGFR-1-deficient ES cells into three dimensional, post-implantation, embryonic tissues, known as embryoid bodies (EBs). We show that the targeted disruption of FGFR-1 leads to a reduced binding of FGF-2 which has no significant effect on the proliferation of undifferentiated ES cells. In addition, lack of functional FGFR-1 in differentiating EBs leads to a reduced expression of the endoderm marker gene alpha-fetoprotein (AFP). This deregulation of the AFP gene correlates with defects in the formation of the visceral endoderm, proper differentiation of the ectoderm and thus the organization of the columnar epithelium, and a block of cavitation. Although the addition of exogenous FGF-2 further reduced the expression of AFPmRNA in differentiating mutant EBs, corresponding morphological changes were not observed. Our results indicate that FGFR-1 may play a vital role in endoderm formation.


Subject(s)
Endoderm/metabolism , Mice/embryology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Animals , Cell Differentiation/physiology , Epithelium/embryology , Ligands , Receptor Protein-Tyrosine Kinases/deficiency , Receptor, Fibroblast Growth Factor, Type 1 , Receptors, Fibroblast Growth Factor/deficiency , Stem Cells/metabolism , alpha-Fetoproteins/biosynthesis , alpha-Fetoproteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...