Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Sci Adv ; 10(12): eadl0788, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38517962

ABSTRACT

The transcription factor FOXL2 is required in ovarian somatic cells for female fertility. Differential timing of Foxl2 deletion, in embryonic versus adult mouse ovary, leads to distinctive outcomes, suggesting different roles across development. Here, we comprehensively investigated FOXL2's role through a multi-omics approach to characterize gene expression dynamics and chromatin accessibility changes, coupled with genome-wide identification of FOXL2 targets and on-chromatin interacting partners in somatic cells across ovarian development. We found that FOXL2 regulates more targets postnatally, through interaction with factors regulating primordial follicle formation and steroidogenesis. Deletion of one interactor, ubiquitin-specific protease 7 (Usp7), results in impairment of somatic cell differentiation, germ cell nest breakdown, and ovarian development, leading to sterility. Our datasets constitute a comprehensive resource for exploration of the molecular mechanisms of ovarian development and causes of female infertility.


Subject(s)
Forkhead Transcription Factors , Ovary , Animals , Mice , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Ovary/metabolism , Ovarian Follicle/metabolism , Gene Expression Regulation , Chromatin/genetics , Chromatin/metabolism
2.
Mol Cell ; 83(18): 3253-3267.e7, 2023 09 21.
Article in English | MEDLINE | ID: mdl-37683646

ABSTRACT

RNA polymerase II (RNAPII) transcription involves initiation from a promoter, transcriptional elongation through the gene, and termination in the terminator region. In bacteria, terminators often contain specific DNA elements provoking polymerase dissociation, but RNAPII transcription termination is thought to be driven entirely by protein co-factors. We used biochemical reconstitution, single-molecule studies, and genome-wide analysis in yeast to study RNAPII termination. Transcription into natural terminators by pure RNAPII results in spontaneous termination at specific sequences containing T-tracts. Single-molecule analysis indicates that termination involves pausing without backtracking. The "torpedo" Rat1-Rai1 exonuclease (XRN2 in humans) greatly stimulates spontaneous termination but is ineffectual on other paused RNAPIIs. By contrast, elongation factor Spt4-Spt5 (DSIF) suppresses termination. Genome-wide analysis further indicates that termination occurs by transcript cleavage at the poly(A) site exposing a new 5' RNA-end that allows Rat1-Rai1 loading, which then catches up with destabilized RNAPII at specific termination sites to end transcription.


Subject(s)
RNA Polymerase II , Saccharomyces cerevisiae Proteins , Humans , RNA Polymerase II/genetics , DNA , Transcription, Genetic , Exonucleases , Peptide Elongation Factors , Saccharomyces cerevisiae/genetics , RNA-Binding Proteins , Saccharomyces cerevisiae Proteins/genetics
3.
Cell Rep Methods ; 3(4): 100456, 2023 04 24.
Article in English | MEDLINE | ID: mdl-37159667

ABSTRACT

Decreased left ventricle (LV) function caused by genetic mutations or injury often leads to debilitating and fatal cardiovascular disease. LV cardiomyocytes are, therefore, a potentially valuable therapeutical target. Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) are neither homogeneous nor functionally mature, which reduces their utility. Here, we exploit cardiac development knowledge to instruct differentiation of hPSCs specifically toward LV cardiomyocytes. Correct mesoderm patterning and retinoic acid pathway blocking are essential to generate near-homogenous LV-specific hPSC-CMs (hPSC-LV-CMs). These cells transit via first heart field progenitors and display typical ventricular action potentials. Importantly, hPSC-LV-CMs exhibit increased metabolism, reduced proliferation, and improved cytoarchitecture and functional maturity compared with age-matched cardiomyocytes generated using the standard WNT-ON/WNT-OFF protocol. Similarly, engineered heart tissues made from hPSC-LV-CMs are better organized, produce higher force, and beat more slowly but can be paced to physiological levels. Together, we show that functionally matured hPSC-LV-CMs can be obtained rapidly without exposure to current maturation regimes.


Subject(s)
Cardiovascular Diseases , Pluripotent Stem Cells , Humans , Myocytes, Cardiac , Heart Ventricles , Action Potentials
4.
Sci Adv ; 9(1): eabn9793, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36598988

ABSTRACT

During embryonic development, mutually antagonistic signaling cascades determine gonadal fate toward a testicular or ovarian identity. Errors in this process result in disorders of sex development (DSDs), characterized by discordance between chromosomal, gonadal, and anatomical sex. The absence of an appropriate, accessible in vitro system is a major obstacle in understanding mechanisms of sex-determination/DSDs. Here, we describe protocols for differentiation of mouse and human pluripotent cells toward gonadal progenitors. Transcriptomic analysis reveals that the in vitro-derived murine gonadal cells are equivalent to embryonic day 11.5 in vivo progenitors. Using similar conditions, Sertoli-like cells derived from 46,XY human induced pluripotent stem cells (hiPSCs) exhibit sustained expression of testis-specific genes, secrete anti-Müllerian hormone, migrate, and form tubular structures. Cells derived from 46,XY DSD female hiPSCs, carrying an NR5A1 variant, show aberrant gene expression and absence of tubule formation. CRISPR-Cas9-mediated variant correction rescued the phenotype. This is a robust tool to understand mechanisms of sex determination and model DSDs.


Subject(s)
Gonadal Dysgenesis, 46,XY , Induced Pluripotent Stem Cells , Male , Animals , Mice , Humans , Female , Cellular Reprogramming/genetics , Gonads , Gonadal Dysgenesis, 46,XY/genetics
5.
J Exp Med ; 220(3)2023 03 06.
Article in English | MEDLINE | ID: mdl-36662229

ABSTRACT

Migration and adhesion play critical roles in B cells, regulating recirculation between lymphoid organs, migration within lymphoid tissue, and interaction with CD4+ T cells. However, there is limited knowledge of how B cells integrate chemokine receptor and integrin signaling with B cell activation to generate efficient humoral responses. Here, we show that the WNK1 kinase, a regulator of migration and adhesion, is essential in B cells for T-dependent and -independent antibody responses. We demonstrate that WNK1 transduces signals from the BCR, CXCR5, and CD40, and using intravital imaging, we show that WNK1 regulates migration of naive and activated B cells, and their interactions with T cells. Unexpectedly, we show that WNK1 is required for BCR- and CD40-induced proliferation, acting through the OXSR1 and STK39 kinases, and for efficient B cell-T cell collaboration in vivo. Thus, WNK1 is critical for humoral immune responses, by regulating B cell migration, adhesion, and T cell-dependent activation.


Subject(s)
Antibody Formation , B-Lymphocytes , Mice , Animals , Lymphoid Tissue , Signal Transduction , CD4-Positive T-Lymphocytes , CD40 Antigens/metabolism , WNK Lysine-Deficient Protein Kinase 1/metabolism
6.
Cancer Cell ; 41(1): 70-87.e14, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36332625

ABSTRACT

The evolution of established cancers is driven by selection of cells with enhanced fitness. Subclonal mutations in numerous epigenetic regulator genes are common across cancer types, yet their functional impact has been unclear. Here, we show that disruption of the epigenetic regulatory network increases the tolerance of cancer cells to unfavorable environments experienced within growing tumors by promoting the emergence of stress-resistant subpopulations. Disruption of epigenetic control does not promote selection of genetically defined subclones or favor a phenotypic switch in response to environmental changes. Instead, it prevents cells from mounting an efficient stress response via modulation of global transcriptional activity. This "transcriptional numbness" lowers the probability of cell death at early stages, increasing the chance of long-term adaptation at the population level. Our findings provide a mechanistic explanation for the widespread selection of subclonal epigenetic-related mutations in cancer and uncover phenotypic inertia as a cellular trait that drives subclone expansion.


Subject(s)
Neoplasms , Humans , Mutation , Neoplasms/genetics , Neoplasms/pathology , Phenotype
7.
Mol Cell Biol ; 42(10): e0017322, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36121223

ABSTRACT

During the heat shock response (HSR), heat shock factor (HSF1 in mammals) binds to target gene promoters, resulting in increased expression of heat shock proteins that help maintain protein homeostasis and ensure cell survival. Besides HSF1, only a relatively few transcription factors with a specific role in ensuring correctly regulated gene expression during the HSR have been described. Here, we use proteomic and genomic (CRISPR) screening to identify a role for RPRD1B in the response to heat shock. Indeed, cells depleted for RPRD1B are heat shock sensitive and show decreased expression of key heat shock proteins (HSPs). These results add to our understanding of the connection between basic gene expression mechanisms and the HSR.


Subject(s)
Heat-Shock Response , Proteomics , Animals , Heat Shock Transcription Factors/genetics , Heat Shock Transcription Factors/metabolism , Heat-Shock Response/genetics , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Mammals/metabolism , HSP70 Heat-Shock Proteins/metabolism
9.
Stem Cell Reports ; 17(4): 894-910, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35334218

ABSTRACT

The spinal cord emerges from a niche of neuromesodermal progenitors (NMPs) formed and maintained by WNT/fibroblast growth factor (FGF) signals at the posterior end of the embryo. NMPs can be generated from human pluripotent stem cells and hold promise for spinal cord replacement therapies. However, NMPs are transient, which compromises production of the full range of rostrocaudal spinal cord identities in vitro. Here we report the generation of NMP-derived pre-neural progenitors (PNPs) with stem cell-like self-renewal capacity. PNPs maintain pre-spinal cord identity for 7-10 passages, dividing to self-renew and to make neural crest progenitors, while gradually adopting a more posterior identity by activating colinear HOX gene expression. The HOX clock can be halted through GDF11-mediated signal inhibition to produce a PNP and NC population with a thoracic identity that can be maintained for up to 30 passages.


Subject(s)
Neural Crest , Pluripotent Stem Cells , Body Patterning/genetics , Bone Morphogenetic Proteins/metabolism , Cell Differentiation/genetics , Fibroblast Growth Factors/metabolism , Growth Differentiation Factors/metabolism , Humans , Neural Crest/metabolism , Pluripotent Stem Cells/metabolism , Spinal Cord/metabolism
10.
Mol Cell ; 82(8): 1573-1588.e10, 2022 04 21.
Article in English | MEDLINE | ID: mdl-35114099

ABSTRACT

The heat shock (HS) response involves rapid induction of HS genes, whereas transcriptional repression is established more slowly at most other genes. Previous data suggested that such repression results from inhibition of RNA polymerase II (RNAPII) pause release, but here, we show that HS strongly affects other phases of the transcription cycle. Intriguingly, while elongation rates increase upon HS, processivity markedly decreases, so that RNAPII frequently fails to reach the end of genes. Indeed, HS results in widespread premature transcript termination at cryptic, intronic polyadenylation (IPA) sites near gene 5'-ends, likely via inhibition of U1 telescripting. This results in dramatic reconfiguration of the human transcriptome with production of new, previously unannotated, short mRNAs that accumulate in the nucleus. Together, these results shed new light on the basic transcription mechanisms induced by growth at elevated temperature and show that a genome-wide shift toward usage of IPA sites can occur under physiological conditions.


Subject(s)
Polyadenylation , Transcriptome , Heat-Shock Response/genetics , Humans , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA, Messenger/genetics
11.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35042798

ABSTRACT

Mutations in the SETX gene, which encodes Senataxin, are associated with the progressive neurodegenerative diseases ataxia with oculomotor apraxia 2 (AOA2) and amyotrophic lateral sclerosis 4 (ALS4). To identify the causal defect in AOA2, patient-derived cells and SETX knockouts (human and mouse) were analyzed using integrated genomic and transcriptomic approaches. A genome-wide increase in chromosome instability (gains and losses) within genes and at chromosome fragile sites was observed, resulting in changes to gene-expression profiles. Transcription stress near promoters correlated with high GCskew and the accumulation of R-loops at promoter-proximal regions, which localized with chromosomal regions where gains and losses were observed. In the absence of Senataxin, the Cockayne syndrome protein CSB was required for the recruitment of the transcription-coupled repair endonucleases (XPG and XPF) and RAD52 recombination protein to target and resolve transcription bubbles containing R-loops, leading to genomic instability. These results show that transcription stress is an important contributor to SETX mutation-associated chromosome fragility and AOA2.


Subject(s)
Chromosomal Instability/genetics , DNA Helicases/metabolism , Multifunctional Enzymes/metabolism , RNA Helicases/metabolism , Spinocerebellar Ataxias/congenital , Animals , Apraxias/genetics , Ataxia/genetics , Cell Line , Cerebellar Ataxia/genetics , DNA Helicases/genetics , DNA Repair/genetics , Gene Expression Profiling/methods , Genomic Instability/genetics , Genomics/methods , Humans , Mice , Mouse Embryonic Stem Cells , Multifunctional Enzymes/genetics , Mutation/genetics , Neurodegenerative Diseases/genetics , Primary Cell Culture , Promoter Regions, Genetic/genetics , RNA Helicases/genetics , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/physiopathology , Transcriptome/genetics
12.
Cell Rep Methods ; 2(12): 100368, 2022 12 19.
Article in English | MEDLINE | ID: mdl-36590686

ABSTRACT

Transcription of protein-coding genes is regulated by dynamic association of co-factors with RNA polymerase II (RNAPII). The function of these factors and their relationship with RNAPII is often poorly understood. Here, we present an approach for elongation-factor-specific mNET capture (ELCAP) of RNAPII complexes for sequencing and mass spectrometry analysis aimed at investigating the function of such RNAPII regulatory proteins. As proof of principle, we apply ELCAP to the RNAPII-associated proteins SCAF4 and SCAF8, which share an essential role as mRNA anti-terminators but have individual roles at the 3' end of genes. Mass spectrometry analysis shows that both SCAF4 and SCAF8 are part of RNAPII elongation complexes containing 3' end processing factors but depleted of splicing components. Importantly, the ELCAP sequencing (ELCAP-seq) profiles of SCAF4- and SCAF8-RNAPII complexes nicely reflect their function as mRNA-anti-terminators and their competing functions at the end of genes, where they prevent or promote transcriptional readthrough.


Subject(s)
Peptide Elongation Factors , RNA Polymerase II , RNA Polymerase II/genetics , Peptide Elongation Factors/genetics , Transcription Factors/genetics , RNA, Messenger/genetics , RNA Splicing/genetics
14.
Nat Cell Biol ; 23(6): 608-619, 2021 06.
Article in English | MEDLINE | ID: mdl-34108662

ABSTRACT

Correct transcription is crucial for life. However, DNA damage severely impedes elongating RNA polymerase II, causing transcription inhibition and transcription-replication conflicts. Cells are equipped with intricate mechanisms to counteract the severe consequence of these transcription-blocking lesions. However, the exact mechanism and factors involved remain largely unknown. Here, using a genome-wide CRISPR-Cas9 screen, we identified the elongation factor ELOF1 as an important factor in the transcription stress response following DNA damage. We show that ELOF1 has an evolutionarily conserved role in transcription-coupled nucleotide excision repair (TC-NER), where it promotes recruitment of the TC-NER factors UVSSA and TFIIH to efficiently repair transcription-blocking lesions and resume transcription. Additionally, ELOF1 modulates transcription to protect cells against transcription-mediated replication stress, thereby preserving genome stability. Thus, ELOF1 protects the transcription machinery from DNA damage via two distinct mechanisms.


Subject(s)
DNA Damage , DNA Repair , Genomic Instability , Peptide Elongation Factor 1/metabolism , Transcription Elongation, Genetic , CRISPR-Cas Systems , Carrier Proteins/genetics , Carrier Proteins/metabolism , Evolution, Molecular , HCT116 Cells , Humans , Peptide Elongation Factor 1/genetics , RNA Polymerase II/metabolism , Transcription Factor TFIIH/genetics , Transcription Factor TFIIH/metabolism , Ubiquitination
15.
Mol Cell ; 81(13): 2808-2822.e10, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34111399

ABSTRACT

The cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway senses cytosolic DNA and induces interferon-stimulated genes (ISGs) to activate the innate immune system. Here, we report the unexpected discovery that cGAS also senses dysfunctional protein production. Purified ribosomes interact directly with cGAS and stimulate its DNA-dependent activity in vitro. Disruption of the ribosome-associated protein quality control (RQC) pathway, which detects and resolves ribosome collision during translation, results in cGAS-dependent ISG expression and causes re-localization of cGAS from the nucleus to the cytosol. Indeed, cGAS preferentially binds collided ribosomes in vitro, and orthogonal perturbations that result in elevated levels of collided ribosomes and RQC activation cause sub-cellular re-localization of cGAS and ribosome binding in vivo as well. Thus, translation stress potently increases DNA-dependent cGAS activation. These findings have implications for the inflammatory response to viral infection and tumorigenesis, both of which substantially reprogram cellular protein synthesis.


Subject(s)
Cell Nucleus , Nucleotidyltransferases , Protein Biosynthesis , Ribosomes , Signal Transduction , Stress, Physiological , Active Transport, Cell Nucleus , Cell Nucleus/chemistry , Cell Nucleus/genetics , Cell Nucleus/metabolism , HEK293 Cells , Humans , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Ribosomes/chemistry , Ribosomes/genetics , Ribosomes/metabolism
16.
J Exp Med ; 218(2)2021 02 01.
Article in English | MEDLINE | ID: mdl-33119032

ABSTRACT

Memory B cells (MBCs) are long-lived cells that form a critical part of immunological memory, providing rapid antibody responses to recurring infections. However, very little is known about signals controlling MBC survival. Previous work has shown that antigen is not required for MBC survival, but a requirement for the B cell antigen receptor (BCR) has not been tested. Other studies have shown that, unlike naive B cells, MBCs do not express BAFFR and their survival is independent of BAFF, the ligand for BAFFR. Here, using inducible genetic ablation, we show that survival of MBCs is critically dependent on the BCR and on signaling through the associated CD79A protein. Unexpectedly, we found that MBCs express BAFFR and that their survival requires BAFF and BAFFR; hence, loss of BAFF or BAFFR impairs recall responses. Finally, we show that MBC survival requires IKK2, a kinase that transduces BAFFR signals. Thus, MBC survival is critically dependent on signaling from BCR and BAFFR.


Subject(s)
B-Cell Activating Factor/immunology , B-Lymphocytes/immunology , Immunologic Memory/immunology , Receptors, Antigen, B-Cell/immunology , Animals , B-Cell Activation Factor Receptor/immunology , Cell Survival , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Signal Transduction/immunology
17.
Cell ; 181(6): 1395-1405.e11, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32531245

ABSTRACT

STK19 was proposed to be a cancer driver, and recent work by Yin et al. (2019) in Cell suggested that the frequently recurring STK19 D89N substitution represents a gain-of-function change, allowing increased phosphorylation of NRAS to enhance melanocyte transformation. Here we show that the STK19 gene has been incorrectly annotated, and that the expressed protein is 110 amino acids shorter than indicated by current databases. The "cancer driving" STK19 D89N substitution is thus outside the coding region. We also fail to detect evidence of the mutation affecting STK19 expression; instead, it is a UV signature mutation, found in the promoter of other genes as well. Furthermore, STK19 is exclusively nuclear and chromatin-associated, while no evidence for it being a kinase was found. The data in this Matters Arising article raise fundamental questions about the recently proposed role for STK19 in melanoma progression via a function as an NRAS kinase, suggested by Yin et al. (2019) in Cell. See also the response by Yin et al. (2020), published in this issue.


Subject(s)
Melanoma , Neoplasm Recurrence, Local , GTP Phosphohydrolases/metabolism , Genes, ras , Humans , Melanoma/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Nuclear Proteins , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Signal Transduction
18.
Cell ; 180(6): 1245-1261.e21, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32142654

ABSTRACT

In response to transcription-blocking DNA damage, cells orchestrate a multi-pronged reaction, involving transcription-coupled DNA repair, degradation of RNA polymerase II (RNAPII), and genome-wide transcription shutdown. Here, we provide insight into how these responses are connected by the finding that ubiquitylation of RNAPII itself, at a single lysine (RPB1 K1268), is the focal point for DNA-damage-response coordination. K1268 ubiquitylation affects DNA repair and signals RNAPII degradation, essential for surviving genotoxic insult. RNAPII degradation results in a shutdown of transcriptional initiation, in the absence of which cells display dramatic transcriptome alterations. Additionally, regulation of RNAPII stability is central to transcription recovery-persistent RNAPII depletion underlies the failure of this process in Cockayne syndrome B cells. These data expose regulation of global RNAPII levels as integral to the cellular DNA-damage response and open the intriguing possibility that RNAPII pool size generally affects cell-specific transcription programs in genome instability disorders and even normal cells.


Subject(s)
DNA Damage , RNA Polymerase II/metabolism , DNA Repair , HEK293 Cells , Humans , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription, Genetic , Ubiquitination , Ultraviolet Rays
19.
Development ; 147(6)2020 03 16.
Article in English | MEDLINE | ID: mdl-32094115

ABSTRACT

Segmentation of the vertebrate hindbrain leads to the formation of rhombomeres, each with a distinct anteroposterior identity. Specialised boundary cells form at segment borders that act as a source or regulator of neuronal differentiation. In zebrafish, there is spatial patterning of neurogenesis in which non-neurogenic zones form at boundaries and segment centres, in part mediated by Fgf20 signalling. To further understand the control of neurogenesis, we have carried out single cell RNA sequencing of the zebrafish hindbrain at three different stages of patterning. Analyses of the data reveal known and novel markers of distinct hindbrain segments, of cell types along the dorsoventral axis, and of the transition of progenitors to neuronal differentiation. We find major shifts in the transcriptome of progenitors and of differentiating cells between the different stages analysed. Supervised clustering with markers of boundary cells and segment centres, together with RNA-seq analysis of Fgf-regulated genes, has revealed new candidate regulators of cell differentiation in the hindbrain. These data provide a valuable resource for functional investigations of the patterning of neurogenesis and the transition of progenitors to neuronal differentiation.


Subject(s)
Body Patterning/genetics , Rhombencephalon/embryology , Rhombencephalon/metabolism , Transcriptome/physiology , Zebrafish , Animals , Animals, Genetically Modified , Atlases as Topic , Cell Differentiation/genetics , Embryo, Nonmammalian , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/genetics , Neurons/cytology , Neurons/physiology , Single-Cell Analysis/methods , Tissue Distribution , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
20.
Biol Open ; 9(2)2020 02 11.
Article in English | MEDLINE | ID: mdl-31988092

ABSTRACT

During vertebrate embryonic development, the formation of axial structures is driven by a population of stem-like cells that reside in a region of the tailbud called the chordoneural hinge (CNH). We have compared the mouse CNH transcriptome with those of surrounding tissues and shown that the CNH and tailbud mesoderm are transcriptionally similar, and distinct from the presomitic mesoderm. Amongst CNH-enriched genes are several that are required for axial elongation, including Wnt3a, Cdx2, Brachyury/T and Fgf8, and androgen/oestrogen receptor nuclear signalling components such as Greb1 We show that the pattern and duration of tailbud Greb1 expression is conserved in mouse, zebrafish and chicken embryos, and that Greb1 is required for axial elongation and somitogenesis in zebrafish embryos. The axial truncation phenotype of Greb1 morphant embryos can be explained by much reduced expression of No tail (Ntl/Brachyury), which is required for axial progenitor maintenance. Posterior segmentation defects in the morphants (including misexpression of genes such as mespb, myoD and papC) appear to result, in part, from lost expression of the segmentation clock gene, her7.


Subject(s)
Embryonic Development/genetics , Membrane Proteins/genetics , Morphogenesis/genetics , Neoplasm Proteins/genetics , Zebrafish Proteins/genetics , Animals , Computational Biology/methods , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Mice , Phenotype , Stem Cells/cytology , Stem Cells/metabolism , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...