Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Development ; 149(22)2022 11 15.
Article in English | MEDLINE | ID: mdl-36326003

ABSTRACT

Stem cell-derived three-dimensional (3D) gastruloids show a remarkable capacity of self-organisation and recapitulate many aspects of gastrulation stage mammalian development. Gastruloids can be rapidly generated and offer several experimental advantages, such as scalability, observability and accessibility for manipulation. Here, we present approaches to further expand the experimental potency of murine 3D gastruloids by using functional genetics in mouse embryonic stem cells (mESCs) to generate chimeric gastruloids. In chimeric gastruloids, fluorescently labelled cells of different genotypes harbouring inducible gene expression or loss-of-function alleles are combined with wild-type cells. We showcase this experimental approach in chimeric gastruloids of mESCs carrying homozygous deletions of the Tbx transcription factor brachyury or inducible expression of Eomes. Resulting chimeric gastruloids recapitulate reported Eomes and brachyury functions, such as instructing cardiac fate and promoting posterior axial extension, respectively. Additionally, chimeric gastruloids revealed previously unrecognised phenotypes, such as the tissue sorting preference of brachyury deficient cells to endoderm and the cell non-autonomous effects of brachyury deficiency on Wnt3a patterning along the embryonic axis, demonstrating some of the advantages of chimeric gastruloids as an efficient tool for studies of mammalian gastrulation.


Subject(s)
Gastrulation , Mammals , Animals , Mice , Endoderm , Mouse Embryonic Stem Cells , Alleles
2.
Development ; 148(1)2021 01 07.
Article in English | MEDLINE | ID: mdl-33199445

ABSTRACT

Anterior mesoderm (AM) and definitive endoderm (DE) progenitors represent the earliest embryonic cell types that are specified during germ layer formation at the primitive streak (PS) of the mouse embryo. Genetic experiments indicate that both lineages segregate from Eomes-expressing progenitors in response to different Nodal signaling levels. However, the precise spatiotemporal pattern of the emergence of these cell types and molecular details of lineage segregation remain unexplored. We combined genetic fate labeling and imaging approaches with single-cell RNA sequencing (scRNA-seq) to follow the transcriptional identities and define lineage trajectories of Eomes-dependent cell types. Accordingly, all cells moving through the PS during the first day of gastrulation express Eomes AM and DE specification occurs before cells leave the PS from Eomes-positive progenitors in a distinct spatiotemporal pattern. ScRNA-seq analysis further suggested the immediate and complete separation of AM and DE lineages from Eomes-expressing cells as last common bipotential progenitor.


Subject(s)
Cell Lineage , Endoderm/cytology , Endoderm/metabolism , Gastrulation , Mesoderm/cytology , Mesoderm/metabolism , Alleles , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Germ Layers/cytology , Mice , Models, Biological , Primitive Streak/embryology , Primitive Streak/metabolism , Stem Cells/metabolism , T-Box Domain Proteins/metabolism , Time Factors , Transcription, Genetic
3.
Genesis ; 55(8)2017 08.
Article in English | MEDLINE | ID: mdl-28646547

ABSTRACT

The T-box transcription factor Eomes (also known as Tbr2) shows short-lived expression in various localized domains of the embryo, including epiblast cells during gastrulation and intermediate progenitor cells in the cerebral cortex. In these tissues Eomes fulfills crucial roles for lineage specification of progenitors. To directly observe Eomes-dependent cell lineages in the living embryo, we generated a novel dual-fluorescence reporter allele that expresses a membrane-bound tdTomato protein for investigation of cell morphology and a nuclear GFP for cell tracing. This allele recapitulates endogenous EOMES protein expression and is suitable for live imaging. We found that the allele can also be used as a short-to-medium-term lineage tracer, as GFP persists in cells longer than EOMES protein and marks Eomes-dependent lineages with a timeframe of days to weeks depending on the proliferation rate. In summary, we present a novel genetic tool for investigation of Eomes-dependent cell types by live imaging and lineage tracing.


Subject(s)
Cell Lineage , Cell Tracking/methods , Genes, Reporter , Genetic Engineering/methods , Optical Imaging/methods , T-Box Domain Proteins/genetics , Animals , Female , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , T-Box Domain Proteins/metabolism
4.
Development ; 138(10): 1913-23, 2011 May.
Article in English | MEDLINE | ID: mdl-21471156

ABSTRACT

The essential roles of SHH in anteroposterior (AP) and AER-FGF signalling in proximodistal (PD) limb bud development are well understood. In addition, these morphoregulatory signals are key components of the self-regulatory SHH/GREM1/AER-FGF feedback signalling system that regulates distal progression of limb bud development. This study uncovers an additional signalling module required for coordinated progression of limb bud axis development. Transcriptome analysis using Shh-deficient mouse limb buds revealed that the expression of proximal genes was distally extended from early stages onwards, which pointed to a more prominent involvement of SHH in PD limb axis development. In particular, retinoic acid (RA) target genes were upregulated proximally, while the expression of the RA-inactivating Cyp26b1 enzyme was downregulated distally, pointing to increased RA activity in Shh-deficient mouse limb buds. Further genetic and molecular analysis established that Cyp26b1 expression is regulated by AER-FGF signalling. During initiation of limb bud outgrowth, the activation of Cyp26b1 expression creates a distal 'RA-free' domain, as indicated by complementary downregulation of a transcriptional sensor of RA activity. Subsequently, Cyp26b1 expression increases as a consequence of SHH-dependent upregulation of AER-FGF signalling. To better understand the underlying signalling interactions, computational simulations of the spatiotemporal expression patterns and interactions were generated. These simulations predicted the existence of an antagonistic AER-FGF/CYP26B1/RA signalling module, which was verified experimentally. In summary, SHH promotes distal progression of limb development by enhancing CYP26B1-mediated RA clearance as part of a signalling network linking the SHH/GREM1/AER-FGF feedback loop to the newly identified AER-FGF/CYP26B1/RA module.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Fibroblast Growth Factors/metabolism , Hedgehog Proteins/metabolism , Limb Buds/embryology , Limb Buds/metabolism , Tretinoin/metabolism , Animals , Cytochrome P-450 Enzyme System/genetics , Ectoderm/embryology , Ectoderm/metabolism , Enzyme Activation , Feedback, Physiological , Female , Fibroblast Growth Factors/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Mesoderm/embryology , Mesoderm/metabolism , Mice , Mice, Knockout , Mice, Mutant Strains , Mutation , Oligonucleotide Array Sequence Analysis , Pregnancy , Retinoic Acid 4-Hydroxylase , Signal Transduction
5.
Differentiation ; 85(4-5): 121-30, 2013.
Article in English | MEDLINE | ID: mdl-23792766

ABSTRACT

Endochondral bone development is orchestrated by the spatially and temporally coordinated differentiation of chondrocytes along the longitudinal axis of the cartilage anlage. Initially, the slowly proliferating, periarticular chondrocytes give rise to the pool of rapidly dividing columnar chondrocytes, whose expansion determines the length of the long bones. The Indian hedgehog (IHH) ligand regulates both the proliferation of columnar chondrocytes and their differentiation into post-mitotic hypertrophic chondrocytes in concert with GLI3, one of the main transcriptional effectors of HH signal transduction. In the absence of Hh signalling, the expression of Vlk (vertebrate lonesome kinase, also called Pkdcc) is increased. We now show that the shortening of limb long bones in Vlk-deficient mouse embryos is aggravated by additional inactivation of Gli3. Our analysis establishes that Vlk and Gli3 synergize to control the temporal kinetics of chondrocyte differentiation during long bone development. Whereas differentiation of limb mesenchymal progenitors into chondrocytes and the initial formation of the cartilage anlagen of the limb skeleton are not altered, Vlk and Gli3 are required for the temporally coordinated differentiation of periarticular into columnar and ultimately hypertrophic chondrocytes in long bones. In limbs lacking both Vlk and Gli3, the appearance of columnar and hypertrophic chondrocytes is severely delayed and zones of morphologically distinct chondrocytes are not established until E16.5. At the molecular level, these morphological alterations are reflected by delayed activation and lowered expression of Ihh, Pth1r and Col10a1 in long bone rudiments of double mutant limbs. In summary, our genetic analysis establishes that VLK plays a role in the IHH/GLI3 interactions and that Vlk and Gli3 cooperate to regulate long bone development by modulating the temporal kinetics of establishing columnar and hypertrophic chondrocyte domains.


Subject(s)
Bone Development/genetics , Bone and Bones/metabolism , Cell Differentiation/genetics , Chondrocytes/metabolism , Kruppel-Like Transcription Factors/genetics , Nerve Tissue Proteins/genetics , Protein Kinases/genetics , Animals , Bone Development/physiology , Bone and Bones/cytology , Bone and Bones/embryology , Cartilage/metabolism , Cell Proliferation , Chondrocytes/cytology , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Parathyroid Hormone-Related Protein/genetics , Parathyroid Hormone-Related Protein/metabolism , Protein-Tyrosine Kinases , Signal Transduction/physiology , Zinc Finger Protein Gli3
6.
Biochim Biophys Acta Mol Cell Res ; 1871(4): 119686, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38342310

ABSTRACT

BACKGROUND: Understanding the genetic underpinnings of protein networks conferring stemness is of broad interest for basic and translational research. METHODS: We used multi-omics analyses to identify and characterize stemness genes, and focused on the zinc finger protein 982 (Zfp982) that regulates stemness through the expression of Nanog, Zfp42, and Dppa3 in mouse embryonic stem cells (mESC). RESULTS: Zfp982 was expressed in stem cells, and bound to chromatin through a GCAGAGKC motif, for example near the stemness genes Nanog, Zfp42, and Dppa3. Nanog and Zfp42 were direct targets of ZFP982 that decreased in expression upon knockdown and increased upon overexpression of Zfp982. We show that ZFP982 expression strongly correlated with stem cell characteristics, both on the transcriptional and morphological levels. Zfp982 expression decreased with progressive differentiation into ecto-, endo- and mesodermal cell lineages, and knockdown of Zfp982 correlated with morphological and transcriptional features of differentiated cells. Zfp982 showed transcriptional overlap with members of the Hippo signaling pathway, one of which was Yap1, the major co-activator of Hippo signaling. Despite the observation that ZFP982 and YAP1 interacted and localized predominantly to the cytoplasm upon differentiation, the localization of YAP1 was not influenced by ZFP982 localization. CONCLUSIONS: Together, our study identified ZFP982 as a transcriptional regulator of early stemness genes, and since ZFP982 is under the control of the Hippo pathway, underscored the importance of the context-dependent Hippo signals for stem cell characteristics.


Subject(s)
Mouse Embryonic Stem Cells , Transcription Factors , Animals , Mice , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation/genetics , Chromosomal Proteins, Non-Histone/metabolism , Mouse Embryonic Stem Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism
7.
Dev Cell ; 58(18): 1627-1642.e7, 2023 09 25.
Article in English | MEDLINE | ID: mdl-37633271

ABSTRACT

Mammalian specification of mesoderm and definitive endoderm (DE) is instructed by the two related Tbx transcription factors (TFs) Eomesodermin (Eomes) and Brachyury sharing partially redundant functions. Gross differences in mutant embryonic phenotypes suggest specific functions of each TF. To date, the molecular details of separated lineage-specific gene regulation by Eomes and Brachyury remain poorly understood. Here, we combine mouse embryonic and stem-cell-based analyses to delineate the non-overlapping, lineage-specific transcriptional activities. On a genome-wide scale, binding of both TFs overlaps at promoters of target genes but shows specificity for distal enhancer regions that is conferred by differences in Tbx DNA-binding motifs. The unique binding to enhancer sites instructs the specification of anterior mesoderm (AM) and DE by Eomes and caudal mesoderm by Brachyury. Remarkably, EOMES antagonizes BRACHYURY gene regulatory functions in coexpressing cells during early gastrulation to ensure the proper sequence of early AM and DE lineage specification followed by posterior mesoderm derivatives.


Subject(s)
Gastrulation , T-Box Domain Proteins , Mice , Animals , Gastrulation/genetics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Mesoderm/metabolism , Fetal Proteins/genetics , Fetal Proteins/metabolism , Gene Expression Regulation, Developmental , Mammals/metabolism
8.
Dev Cell ; 56(23): 3276-3287.e8, 2021 12 06.
Article in English | MEDLINE | ID: mdl-34741805

ABSTRACT

The process of implantation and the cellular interactions at the embryo-maternal interface are intrinsically difficult to analyze, as the implanting embryo is concealed by the uterine tissues. Therefore, the mechanisms mediating the interconnection of the embryo and the mother are poorly understood. Here, we established a 3D biomimetic culture environment that harbors the key features of the murine implantation niche. This culture system enabled direct analysis of trophoblast invasion and revealed the first embryonic interactions with the maternal vasculature. We found that implantation is mediated by the collective migration of penetrating strands of trophoblast giant cells, which acquire the expression of vascular receptors, ligands, and adhesion molecules, assembling a network for communication with the maternal blood vessels. In particular, Pdgf signaling cues promote the establishment of the heterologous contacts. Together, the biomimetic platform and our findings thereof elucidate the hidden dynamics of the early interactions at the implantation site.


Subject(s)
Blastocyst/metabolism , Blood Vessels/metabolism , Cell Communication , Embryo, Mammalian/metabolism , Embryonic Development , Maternal-Fetal Exchange , Trophoblasts/metabolism , Animals , Biomimetics , Blastocyst/cytology , Blood Vessels/cytology , Cell Culture Techniques , Cell Movement , Embryo Implantation , Embryo, Mammalian/cytology , Female , Giant Cells/cytology , Giant Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Pregnancy , Trophoblasts/cytology
9.
Int J Cancer ; 126(5): 1256-62, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-19697324

ABSTRACT

The incidence of human papilloma virus (HPV) induced oropharyngeal squamous cell carcinoma (OPSCC) increases in the western countries. These OPSCC show distinct molecular characteristics and are characterized by an overexpression of p16, considered a surrogate marker for HPV infection. When compared to patients with p16 negative OPSCC, patients with HPV induced p16 positive OPSCC show a significantly better prognosis, which is reported to be caused by increased radiosensitivity. The objective of the present study was to analyze the impact of p16 expression status on the prognosis of OPSCC treated by either radiotherapy (RT) or primary surgery. Results are based upon a tissue microarray (TMA) of 365 head neck squamous cell carcinomas (HNSCC) including 85 OPSCC with clinico-pathological and follow-up data. p16 positivity correlated significantly with oropharyngeal tumor localization (p < 0.001). Patients with p16 positive OPSCC exhibited a significantly better overall survival than those with p16 negative tumors (p = 0.007). In a multivariate analysis, survival benefit of patients with p16 positive OPSCC was independent of clinico-pathological parameters such as cT and cN classification and treatment modality. The improved prognosis of p16 positive OPSCC is found after RT as well as after surgery.


Subject(s)
Carcinoma, Squamous Cell/virology , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Oropharyngeal Neoplasms/virology , Papillomavirus Infections/complications , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Combined Modality Therapy , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Neoplasm Staging , Oropharyngeal Neoplasms/pathology , Oropharyngeal Neoplasms/therapy , Prognosis , Radiotherapy , Tissue Array Analysis
11.
Nat Cell Biol ; 21(12): 1518-1531, 2019 12.
Article in English | MEDLINE | ID: mdl-31792383

ABSTRACT

The first lineage specification of pluripotent mouse epiblast segregates neuroectoderm (NE) from mesoderm and definitive endoderm (ME) by mechanisms that are not well understood. Here we demonstrate that the induction of ME gene programs critically relies on the T-box transcription factors Eomesodermin (also known as Eomes) and Brachyury, which concomitantly repress pluripotency and NE gene programs. Cells deficient in these T-box transcription factors retain pluripotency and differentiate to NE lineages despite the presence of ME-inducing signals transforming growth factor ß (TGF-ß)/Nodal and Wnt. Pluripotency and NE gene networks are additionally repressed by ME factors downstream of T-box factor induction, demonstrating a redundancy in program regulation to safeguard mutually exclusive lineage specification. Analyses of chromatin revealed that accessibility of ME enhancers depends on T-box factor binding, whereas NE enhancers are accessible and already activation primed at pluripotency. This asymmetry of the chromatin landscape thus explains the default differentiation of pluripotent cells to NE in the absence of ME induction that depends on activating and repressive functions of Eomes and Brachyury.


Subject(s)
Chromatin/genetics , Fetal Proteins/genetics , Germ Layers/physiology , Pluripotent Stem Cells/physiology , T-Box Domain Proteins/genetics , Animals , Cell Differentiation/genetics , Cell Line , Cell Separation/methods , Endoderm/physiology , Female , Gene Expression Regulation, Developmental/genetics , Male , Mice , Neural Plate/physiology , Transforming Growth Factor beta/genetics
12.
PLoS One ; 10(8): e0136566, 2015.
Article in English | MEDLINE | ID: mdl-26305214

ABSTRACT

Studies of mammalian tissue culture cells indicate that the conserved and distinct NDR isoforms, NDR1 and NDR2, play essential cell biological roles. However, mice lacking either Ndr1 or Ndr2 alone develop normally. Here, we studied the physiological consequences of inactivating both NDR1 and NDR2 in mice, showing that the lack of both Ndr1/Ndr2 (called Ndr1/2-double null mutants) causes embryonic lethality. In support of compensatory roles for NDR1 and NDR2, total protein and activating phosphorylation levels of the remaining NDR isoform were elevated in mice lacking either Ndr1 or Ndr2. Mice retaining one single wild-type Ndr allele were viable and fertile. Ndr1/2-double null embryos displayed multiple phenotypes causing a developmental delay from embryonic day E8.5 onwards. While NDR kinases are not required for notochord formation, the somites of Ndr1/2-double null embryos were smaller, irregularly shaped and unevenly spaced along the anterior-posterior axis. Genes implicated in somitogenesis were down-regulated and the normally symmetric expression of Lunatic fringe, a component of the Notch pathway, showed a left-right bias in the last forming somite in 50% of all Ndr1/2-double null embryos. In addition, Ndr1/2-double null embryos developed a heart defect that manifests itself as pericardial edemas, obstructed heart tubes and arrest of cardiac looping. The resulting cardiac insufficiency is the likely cause of the lethality of Ndr1/2-double null embryos around E10. Taken together, we show that NDR kinases compensate for each other in vivo in mouse embryos, explaining why mice deficient for either Ndr1 or Ndr2 are viable. Ndr1/2-double null embryos show defects in somitogenesis and cardiac looping, which reveals their essential functions and shows that the NDR kinases are critically required during the early phase of organogenesis.


Subject(s)
Organogenesis/genetics , Protein Serine-Threonine Kinases/genetics , Proteins/genetics , Adaptor Proteins, Signal Transducing , Animals , Embryonic Development/genetics , Humans , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/biosynthesis , Signal Transduction
13.
Wiley Interdiscip Rev Dev Biol ; 1(6): 803-22, 2012.
Article in English | MEDLINE | ID: mdl-23799625

ABSTRACT

This review focuses predominantly on the human congenital malformations caused by alterations affecting the morphoregulatory gene networks that control early limb bud patterning and outgrowth. Limb defects are among the most frequent congenital malformations in humans that are caused by genetic mutations or teratogenic effects resulting either in abnormal, loss of, or additional skeletal elements. Spontaneous and engineered mouse models have been used to identify and study the molecular alterations and disrupted gene networks that underlie human congenital limb malformations. More recently, mouse genetics has begun to reveal the alterations that affect the often-large cis-regulatory landscapes that control gene expression in limb buds and cause devastating effects on limb bud development. These findings have paved the way to identifying mutations in cis-regulatory regions as causal to an increasing number of congenital limb malformations in humans. In these cases, no mutations in the coding region of a presumed candidate were previously detected. This review highlights how the current understanding of the molecular gene networks and interactions that control mouse limb bud development provides insight into the etiology of human congenital limb malformations.


Subject(s)
Limb Deformities, Congenital/genetics , Animals , Disease Models, Animal , Gene Regulatory Networks , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Limb Deformities, Congenital/metabolism , Patched Receptors , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Signal Transduction , Teratogens/metabolism
14.
Hum Pathol ; 39(10): 1527-34, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18620726

ABSTRACT

Epidermal growth factor receptor is overexpressed in more than 80% of head-neck squamous cell carcinoma. Its role as an independent prognostic marker is discussed controversially. No standardized evaluation methods are reported. The aim of our study was to analyze the prognostic relevance of epidermal growth factor receptor expression, using a tissue microarray with more than 300 tumor samples. Epidermal growth factor receptor expression was analyzed by immunohistochemistry and fluorescence in situ hybridization based on a tissue microarray of 365 head-neck squamous cell carcinomas with complete clinicopathologic and follow-up data. Multiple independent observers blinded for clinical data evaluated epidermal growth factor receptor immunostaining semiquantitatively. Cut-off scores for positivity were determined systematically by receiver operating characteristic curve analysis and validated by resampling of the data. Epidermal growth factor receptor expression cut-off scores for loco-regional relapse and overall survival were determined to be 60%. No significant correlation with clinicopathologic data was found. Independent significant differences in loco-regional control and overall survival could not be distinguished by epidermal growth factor receptor expression. Epidermal growth factor receptor expression could not be confirmed as a significant independent prognostic marker in head-neck squamous cell carcinoma using a large tissue microarray with 365 head-neck squamous cell carcinomas with complete clinical data, an evaluation based on immunohistochemistry and fluorescence in situ hybridization by multiple independent observers and systematic determination of cut-off scores.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/metabolism , ErbB Receptors/metabolism , Head and Neck Neoplasms/metabolism , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/mortality , DNA, Neoplasm/analysis , Diagnostic Errors , Female , Fluorescent Antibody Technique, Indirect , Head and Neck Neoplasms/diagnosis , Head and Neck Neoplasms/mortality , Humans , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Male , Prognosis , ROC Curve , Retrospective Studies , Survival Rate , Tissue Array Analysis
SELECTION OF CITATIONS
SEARCH DETAIL