Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Brain ; 2024 May 16.
Article in English | MEDLINE | ID: mdl-38753057

ABSTRACT

Deubiquitination is critical for the proper functioning of numerous biological pathways such as DNA repair, cell cycle progression, transcription, signal transduction, and autophagy. Accordingly, pathogenic variants in deubiquitinating enzymes (DUBs) have been implicated in neurodevelopmental disorders (ND) and congenital abnormalities. ATXN7L3 is a component of the DUB module of the SAGA complex, and two other related DUB modules, and serves as an obligate adaptor protein of 3 ubiquitin-specific proteases (USP22, USP27X or USP51). Through exome sequencing and GeneMatching, we identified nine individuals with heterozygous variants in ATXN7L3. The core phenotype included global motor and language developmental delay, hypotonia, and distinctive facial characteristics including hypertelorism, epicanthal folds, blepharoptosis, a small nose and mouth, and low-set posteriorly rotated ears. In order to assess pathogenicity, we investigated the effects of a recurrent nonsense variant [c.340C>T; p.(Arg114Ter)] in fibroblasts of an affected individual. ATXN7L3 protein levels were reduced, and deubiquitylation was impaired, as indicated by an increase in histone H2Bub1 levels. This is consistent with the previous observation of increased H2Bub1 levels in Atxn7l3-null mouse embryos, which have developmental delay and embryonic lethality. In conclusion, we present clinical information and biochemical characterization supporting ATXN7L3 variants in the pathogenesis of a rare syndromic ND.

2.
bioRxiv ; 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-38076793

ABSTRACT

The recognition of core promoter sequences by the general transcription factor TFIID is the first step in the process of RNA polymerase II (Pol II) transcription initiation. Metazoan holo-TFIID is composed of the TATA binding protein (TBP) and of 13 TBP associated factors (TAFs). Inducible Taf7 knock out (KO) results in the formation of a Taf7-less TFIID complex, while Taf10 KO leads to serious defects within the TFIID assembly pathway. Either TAF7 or TAF10 depletions correlate with the detected TAF occupancy changes at promoters, and with the distinct phenotype severities observed in mouse embryonic stem cells or mouse embryos. Surprisingly however, under either Taf7 or Taf10 deletion conditions, TBP is still associated to the chromatin, and no major changes are observed in nascent Pol II transcription. Thus, partially assembled TFIID complexes can sustain Pol II transcription initiation, but cannot replace holo-TFIID over several cell divisions and/or development.

3.
Cell Rep ; 42(9): 113099, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37682711

ABSTRACT

To understand the function of multisubunit complexes, it is of key importance to uncover the precise mechanisms that guide their assembly. Nascent proteins can find and bind their interaction partners during their translation, leading to co-translational assembly. Here, we demonstrate that the core modules of ATAC (ADA-two-A-containing) and SAGA (Spt-Ada-Gcn5-acetyltransferase), two lysine acetyl transferase-containing transcription co-activator complexes, assemble co-translationally in the cytoplasm of mammalian cells. In addition, a SAGA complex containing all of its modules forms in the cytoplasm and acetylates non-histone proteins. In contrast, ATAC complex subunits cannot be detected in the cytoplasm of mammalian cells. However, an endogenous ATAC complex containing two functional modules forms and functions in the nucleus. Thus, the two related co-activators, ATAC and SAGA, assemble using co-translational pathways, but their subcellular localization, cytoplasmic abundance, and functions are distinct.


Subject(s)
Histone Acetyltransferases , Saccharomyces cerevisiae Proteins , Animals , Histone Acetyltransferases/metabolism , Transcription Factors/metabolism , Chromatin , Cell Nucleus/metabolism , Fungal Proteins , Saccharomyces cerevisiae Proteins/metabolism , Mammals/metabolism
4.
bioRxiv ; 2023 Aug 03.
Article in English | MEDLINE | ID: mdl-37577620

ABSTRACT

To understand the function of multisubunit complexes it is of key importance to uncover the precise mechanisms that guide their assembly. Nascent proteins can find and bind their interaction partners during their translation, leading to co-translational assembly. Here we demonstrate that the core modules of ATAC (ADA-Two-A-Containing) and SAGA (Spt-Ada-Gcn5-acetyltransferase), two lysine acetyl transferase-containing transcription coactivator complexes, assemble co-translationally in the cytoplasm of mammalian cells. In addition, SAGA complex containing all of its modules forms in the cytoplasm and acetylates non-histones proteins. In contrast, fully assembled ATAC complex cannot be detected in the cytoplasm of mammalian cells. However, endogenous ATAC complex containing two functional modules forms and functions in the nucleus. Thus, the two related coactivators, ATAC and SAGA, assemble by using co-translational pathways, but their subcellular localization, cytoplasmic abundance and functions are distinct.

5.
Biomedicines ; 10(10)2022 Oct 20.
Article in English | MEDLINE | ID: mdl-36289911

ABSTRACT

Prdm1 mutant mice are one of the rare mutant strains that do not develop whisker hair follicles while still displaying a pelage. Here, we show that Prdm1 is expressed at the earliest stage of whisker development in clusters of mesenchymal cells before placode formation. Its conditional knockout in the murine soma leads to the loss of expression of Bmp2, Shh, Bmp4, Krt17, Edar, and Gli1, though leaving the ß-catenin-driven first dermal signal intact. Furthermore, we show that Prdm1 expressing cells not only act as a signaling center but also as a multipotent progenitor population contributing to the several lineages of the adult whisker. We confirm by genetic ablation experiments that the absence of macro vibrissae reverberates on the organization of nerve wiring in the mystacial pads and leads to the reorganization of the barrel cortex. We demonstrate that Lef1 acts upstream of Prdm1 and identify a primate-specific deletion of a Lef1 enhancer named Leaf. This loss may have been significant in the evolutionary process, leading to the progressive defunctionalization and disappearance of vibrissae in primates.

6.
Int J Mol Sci ; 23(13)2022 Jul 05.
Article in English | MEDLINE | ID: mdl-35806465

ABSTRACT

Ubiquitin (ub) is a small, highly conserved protein widely expressed in eukaryotic cells. Ubiquitination is a post-translational modification catalyzed by enzymes that activate, conjugate, and ligate ub to proteins. Substrates can be modified either by addition of a single ubiquitin molecule (monoubiquitination), or by conjugation of several ubs (polyubiquitination). Monoubiquitination acts as a signaling mark to control diverse biological processes. The cellular and spatial distribution of ub is determined by the opposing activities of ub ligase enzymes, and deubiquitinases (DUBs), which remove ub from proteins to generate free ub. In mammalian cells, 1-2% of total histone H2B is monoubiquitinated. The SAGA (Spt Ada Gcn5 Acetyl-transferase) is a transcriptional coactivator and its DUB module removes ub from H2Bub1. The mammalian SAGA DUB module has four subunits, ATXN7, ATXN7L3, USP22, and ENY2. Atxn7l3-/- mouse embryos, lacking DUB activity, have a five-fold increase in H2Bub1 retention, and die at mid-gestation. Interestingly, embryos lacking the ub encoding gene, Ubc, have a similar phenotype. Here we provide a current overview of data suggesting that H2Bub1 retention on the chromatin in Atxn7l3-/- embryos may lead to an imbalance in free ub distribution. Thus, we speculate that ATXN7L3-containing DUBs impact the free cellular ub pool during development.


Subject(s)
Histones , Ubiquitin , Animals , Embryonic Development/genetics , Histones/genetics , Histones/metabolism , Mammals/metabolism , Mice , Protein Processing, Post-Translational , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitination
7.
Nucleic Acids Res ; 50(14): 7972-7990, 2022 08 12.
Article in English | MEDLINE | ID: mdl-35871303

ABSTRACT

Coactivator complexes regulate chromatin accessibility and transcription. SAGA (Spt-Ada-Gcn5 Acetyltransferase) is an evolutionary conserved coactivator complex. The core module scaffolds the entire SAGA complex and adopts a histone octamer-like structure, which consists of six histone-fold domain (HFD)-containing proteins forming three histone-fold (HF) pairs, to which the double HFD-containing SUPT3H adds one HF pair. Spt3, the yeast ortholog of SUPT3H, interacts genetically and biochemically with the TATA binding protein (TBP) and contributes to global RNA polymerase II (Pol II) transcription. Here we demonstrate that (i) SAGA purified from human U2OS or mouse embryonic stem cells (mESC) can assemble without SUPT3H, (ii) SUPT3H is not essential for mESC survival, but required for their growth and self-renewal, and (iii) the loss of SUPT3H from mammalian cells affects the transcription of only a specific subset of genes. Accordingly, in the absence of SUPT3H no major change in TBP accumulation at gene promoters was observed. Thus, SUPT3H is not required for the assembly of SAGA, TBP recruitment, or overall Pol II transcription, but plays a role in mESC growth and self-renewal. Our data further suggest that yeast and mammalian SAGA complexes contribute to transcription regulation by distinct mechanisms.


Subject(s)
RNA Polymerase II , Trans-Activators , Transcription Factors , Animals , DNA-Binding Proteins/genetics , Histone Acetyltransferases/metabolism , Histones/genetics , Histones/metabolism , Humans , Mice , RNA Polymerase II/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcription, Genetic
8.
Biochem Soc Trans ; 49(5): 2051-2062, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34415300

ABSTRACT

In somatic cells, RNA polymerase II (Pol II) transcription initiation starts by the binding of the general transcription factor TFIID, containing the TATA-binding protein (TBP) and 13 TBP-associated factors (TAFs), to core promoters. However, in growing oocytes active Pol II transcription is TFIID/TBP-independent, as during oocyte growth TBP is replaced by its vertebrate-specific paralog TBPL2. TBPL2 does not interact with TAFs, but stably associates with TFIIA. The maternal transcriptome is the population of mRNAs produced and stored in the cytoplasm of growing oocytes. After fertilization, maternal mRNAs are inherited by the zygote from the oocyte. As transcription becomes silent after oocyte growth, these mRNAs are the sole source for active protein translation. They will participate to complete the protein pool required for oocyte terminal differentiation, fertilization and initiation of early development, until reactivation of transcription in the embryo, called zygotic genome activation (ZGA). All these events are controlled by an important reshaping of the maternal transcriptome. This procedure combines cytoplasmic readenylation of stored transcripts, allowing their translation, and different waves of mRNA degradation by deadenylation coupled to decapping, to eliminate transcripts coding for proteins that are no longer required. The reshaping ends after ZGA with an almost total clearance of the maternal transcripts. In the past, the murine maternal transcriptome has received little attention but recent progresses have brought new insights into the regulation of maternal mRNA dynamics in the mouse. This review will address past and recent data on the mechanisms associated with maternal transcriptome dynamic in the mouse.


Subject(s)
Embryonic Development/genetics , Gene Expression Regulation, Developmental , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptome/genetics , Animals , Female , Mice , Nuclear Proteins/metabolism , Oocytes/metabolism , Pregnancy , Promoter Regions, Genetic , RNA Polymerase II/metabolism , RNA Stability , TATA Box Binding Protein-Like Proteins/metabolism , TATA-Box Binding Protein/metabolism , Transcription, Genetic , Zygote/growth & development , Zygote/metabolism
9.
Cell Death Differ ; 28(8): 2385-2403, 2021 08.
Article in English | MEDLINE | ID: mdl-33731875

ABSTRACT

Co-activator complexes dynamically deposit post-translational modifications (PTMs) on histones, or remove them, to regulate chromatin accessibility and/or to create/erase docking surfaces for proteins that recognize histone PTMs. SAGA (Spt-Ada-Gcn5 Acetyltransferase) is an evolutionary conserved multisubunit co-activator complex with modular organization. The deubiquitylation module (DUB) of mammalian SAGA complex is composed of the ubiquitin-specific protease 22 (USP22) and three adaptor proteins, ATXN7, ATXN7L3 and ENY2, which are all needed for the full activity of the USP22 enzyme to remove monoubiquitin (ub1) from histone H2B. Two additional USP22-related ubiquitin hydrolases (called USP27X or USP51) have been described to form alternative DUBs with ATXN7L3 and ENY2, which can also deubiquitylate H2Bub1. Here we report that USP22 and ATXN7L3 are essential for normal embryonic development of mice, however their requirements are not identical during this process, as Atxn7l3-/- embryos show developmental delay already at embryonic day (E) 7.5, while Usp22-/- embryos are normal at this stage, but die at E14.5. Global histone H2Bub1 levels were only slightly affected in Usp22 null embryos, in contrast H2Bub1 levels were strongly increased in Atxn7l3 null embryos and derived cell lines. Our transcriptomic analyses carried out from wild type and Atxn7l3-/- mouse embryonic stem cells (mESCs), or primary mouse embryonic fibroblasts (MEFs) suggest that the ATXN7L3-related DUB activity regulates only a subset of genes in both cell types. However, the gene sets and the extent of their deregulation were different in mESCs and MEFs. Interestingly, the strong increase of H2Bub1 levels observed in the Atxn7l3-/- mESCs, or Atxn7l3-/- MEFs, does not correlate with the modest changes in RNA Polymerase II (Pol II) occupancy and lack of changes in Pol II elongation observed in the two Atxn7l3-/- cellular systems. These observations together indicate that deubiquitylation of histone H2Bub1 does not directly regulate global Pol II transcription elongation.


Subject(s)
Gene Expression/genetics , Histones/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Animals , Mice , Transcription Factors/metabolism , Ubiquitination
10.
Nat Commun ; 11(1): 6439, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33353944

ABSTRACT

During oocyte growth, transcription is required to create RNA and protein reserves to achieve maternal competence. During this period, the general transcription factor TATA binding protein (TBP) is replaced by its paralogue, TBPL2 (TBP2 or TRF3), which is essential for RNA polymerase II transcription. We show that in oocytes TBPL2 does not assemble into a canonical TFIID complex. Our transcript analyses demonstrate that TBPL2 mediates transcription of oocyte-expressed genes, including mRNA survey genes, as well as specific endogenous retroviral elements. Transcription start site (TSS) mapping indicates that TBPL2 has a strong preference for TATA-like motif in core promoters driving sharp TSS selection, in contrast with canonical TBP/TFIID-driven TATA-less promoters that have broader TSS architecture. Thus, we show a role for the TBPL2/TFIIA complex in the establishment of the oocyte transcriptome by using a specific TSS recognition code.


Subject(s)
Nuclear Proteins/metabolism , Oocytes/metabolism , Promoter Regions, Genetic , Transcription Factor TFIIA/metabolism , Transcriptome/genetics , Animals , Animals, Newborn , Female , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Mutation/genetics , NIH 3T3 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , TATA Box , Terminal Repeat Sequences/genetics , Transcription Factor TFIID/metabolism , Transcription, Genetic
11.
Cell Rep ; 29(6): 1410-1418.e6, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31693883

ABSTRACT

Browning induction or transplantation of brown adipose tissue (BAT) or brown/beige adipocytes derived from progenitor or induced pluripotent stem cells (iPSCs) can represent a powerful strategy to treat metabolic diseases. However, our poor understanding of the mechanisms that govern the differentiation and activation of brown adipocytes limits the development of such therapy. Various genetic factors controlling the differentiation of brown adipocytes have been identified, although most studies have been performed using in vitro cultured pre-adipocytes. We investigate here the differentiation of brown adipocytes from adipose progenitors in the mouse embryo. We demonstrate that the formation of multiple lipid droplets (LDs) is initiated within clusters of glycogen, which is degraded through glycophagy to provide the metabolic substrates essential for de novo lipogenesis and LD formation. Therefore, this study uncovers the role of glycogen in the generation of LDs.


Subject(s)
Adipocytes, Brown/metabolism , Adipogenesis/genetics , Adipose Tissue, Brown/metabolism , Embryo, Mammalian/metabolism , Glycogen/metabolism , Lipid Droplets/metabolism , Adipocytes, Brown/ultrastructure , Adipose Tissue, Brown/embryology , Adipose Tissue, Brown/ultrastructure , Animals , Autophagy/drug effects , Autophagy/genetics , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cells, Cultured , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Glycogen/ultrastructure , Humans , Lipid Droplets/ultrastructure , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , PPAR gamma/genetics , PPAR gamma/metabolism , RNA, Small Interfering , Transcriptome
12.
Nat Commun ; 10(1): 1740, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30988355

ABSTRACT

Cells dedicate significant energy to build proteins often organized in multiprotein assemblies with tightly regulated stoichiometries. As genes encoding subunits assembling in a multisubunit complex are dispersed in the genome of eukaryotes, it is unclear how these protein complexes assemble. Here, we show that mammalian nuclear transcription complexes (TFIID, TREX-2 and SAGA) composed of a large number of subunits, but lacking precise architectural details are built co-translationally. We demonstrate that dimerization domains and their positions in the interacting subunits determine the co-translational assembly pathway (simultaneous or sequential). The lack of co-translational interaction can lead to degradation of the partner protein. Thus, protein synthesis and complex assembly are linked in building mammalian multisubunit complexes, suggesting that co-translational assembly is a general principle in mammalian cells to avoid non-specific interactions and protein aggregation. These findings will also advance structural biology by defining endogenous co-translational building blocks in the architecture of multisubunit complexes.


Subject(s)
Protein Multimerization , Protein Subunits/metabolism , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/metabolism , HeLa Cells , Humans , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Protein Domains , Protein Folding , Protein Subunits/chemistry , TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/metabolism
13.
Development ; 144(20): 3808-3818, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28893950

ABSTRACT

During development, tightly regulated gene expression programs control cell fate and patterning. A key regulatory step in eukaryotic transcription is the assembly of the pre-initiation complex (PIC) at promoters. PIC assembly has mainly been studied in vitro, and little is known about its composition during development. In vitro data suggest that TFIID is the general transcription factor that nucleates PIC formation at promoters. Here we show that TAF10, a subunit of TFIID and of the transcriptional co-activator SAGA, is required for the assembly of these complexes in the mouse embryo. We performed Taf10 conditional deletions during mesoderm development and show that Taf10 loss in the presomitic mesoderm (PSM) does not prevent cyclic gene transcription or PSM segmental patterning, whereas lateral plate differentiation is profoundly altered. During this period, global mRNA levels are unchanged in the PSM, with only a minor subset of genes dysregulated. Together, our data strongly suggest that the TAF10-containing canonical TFIID and SAGA complexes are dispensable for early paraxial mesoderm development, arguing against the generic role in transcription proposed for these fully assembled holo-complexes.


Subject(s)
Gene Expression Regulation, Developmental , TATA-Binding Protein Associated Factors/metabolism , Trans-Activators/genetics , Transcription Factor TFIID/genetics , Transcription, Genetic , Animals , Body Patterning , Cell Differentiation , Cell Nucleus/metabolism , Gene Deletion , Mesoderm/embryology , Mesoderm/metabolism , Mice , Promoter Regions, Genetic , Protein Binding , Protein Domains , RNA, Messenger/metabolism , TATA-Binding Protein Associated Factors/genetics , Trans-Activators/metabolism , Transcription Factor TFIID/metabolism
14.
Development ; 143(5): 872-9, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26839363

ABSTRACT

Pax3 and Foxc2 have been shown genetically to mutually repress each other in the mouse somite. Perturbation of this balance in multipotent cells of the dermomyotome influences cell fate; upregulation of Foxc2 favours a vascular fate, whereas higher levels of Pax3 lead to myogenesis. Foxc1 has overlapping functions with Foxc2. In Foxc1/2 double-mutant embryos, somitogenesis is severely affected, precluding analysis of somite derivatives. We have adopted a conditional approach whereby mutations in Foxc1 and Foxc2 genes were targeted to Pax3-expressing cells. Inclusion of a conditional reporter allele in the crosses made it possible to follow cells that had expressed Pax3. At the forelimb level, endothelial and myogenic cells migrate from adjacent somites into the limb bud. This population of endothelial cells is compromised in the double mutant, whereas excessive production of myogenic cells is observed in the trunk. However, strikingly, myogenic progenitors fail to enter the limbs, leading to the absence of skeletal muscle. Pax3-positive migratory myogenic progenitors, marked by expression of Lbx1, are specified in the somite at forelimb level, but endothelial progenitors are absent. The myogenic progenitors do not die, but differentiate prematurely adjacent to the somite. We conclude that the small proportion of somite-derived endothelial cells in the limb is required for the migration of myogenic limb progenitors.


Subject(s)
Endothelial Cells/metabolism , Forelimb/embryology , Forkhead Transcription Factors/genetics , Muscle Development/physiology , Paired Box Transcription Factors/metabolism , Somites/metabolism , Animals , Cell Movement , Cell Separation , Crosses, Genetic , Female , Flow Cytometry , Forelimb/metabolism , Gene Expression Regulation, Developmental , Genes, Reporter , In Situ Hybridization , Limb Buds/embryology , Male , Mice , Mice, Transgenic , Muscle Proteins/genetics , Mutation , PAX3 Transcription Factor , Paired Box Transcription Factors/genetics , Phenotype
15.
Genes Dev ; 28(18): 1999-2012, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25228644

ABSTRACT

The SAGA (Spt-Ada-Gcn5 acetyltransferase) coactivator complex contains distinct chromatin-modifying activities and is recruited by DNA-bound activators to regulate the expression of a subset of genes. Surprisingly, recent studies revealed little overlap between genome-wide SAGA-binding profiles and changes in gene expression upon depletion of subunits of the complex. As indicators of SAGA recruitment on chromatin, we monitored in yeast and human cells the genome-wide distribution of histone H3K9 acetylation and H2B ubiquitination, which are respectively deposited or removed by SAGA. Changes in these modifications after inactivation of the corresponding enzyme revealed that SAGA acetylates the promoters and deubiquitinates the transcribed region of all expressed genes. In agreement with this broad distribution, we show that SAGA plays a critical role for RNA polymerase II recruitment at all expressed genes. In addition, through quantification of newly synthesized RNA, we demonstrated that SAGA inactivation induced a strong decrease of mRNA synthesis at all tested genes. Analysis of the SAGA deubiquitination activity further revealed that SAGA acts on the whole transcribed genome in a very fast manner, indicating a highly dynamic association of the complex with chromatin. Thus, our study uncovers a new function for SAGA as a bone fide cofactor for all RNA polymerase II transcription.


Subject(s)
Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation , RNA Polymerase II/genetics , Saccharomyces cerevisiae Proteins/metabolism , Trans-Activators/metabolism , Acetylation , Animals , Gene Expression Profiling , Genome , HEK293 Cells , HeLa Cells , Histones/metabolism , Humans , Mice , Promoter Regions, Genetic , Protein Binding , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Trans-Activators/genetics , Ubiquitination
16.
Proc Natl Acad Sci U S A ; 111(24): 8844-9, 2014 Jun 17.
Article in English | MEDLINE | ID: mdl-24927569

ABSTRACT

Multipotent Pax3-positive (Pax3(+)) cells in the somites give rise to skeletal muscle and to cells of the vasculature. We had previously proposed that this cell-fate choice depends on the equilibrium between Pax3 and Foxc2 expression. In this study, we report that the Notch pathway promotes vascular versus skeletal muscle cell fates. Overactivating the Notch pathway specifically in Pax3(+) progenitors, via a conditional Pax3(NICD) allele, results in an increase of the number of smooth muscle and endothelial cells contributing to the aorta. At limb level, Pax3(+) cells in the somite give rise to skeletal muscles and to a subpopulation of endothelial cells in blood vessels of the limb. We now demonstrate that in addition to the inhibitory role of Notch signaling on skeletal muscle cell differentiation, the Notch pathway affects the Pax3:Foxc2 balance and promotes the endothelial versus myogenic cell fate, before migration to the limb, in multipotent Pax3(+) cells in the somite of the mouse embryo.


Subject(s)
Endothelial Cells/cytology , Extremities/embryology , Gene Expression Regulation, Developmental , Paired Box Transcription Factors/genetics , Receptors, Notch/metabolism , Somites/embryology , Alleles , Animals , Cell Differentiation , Cell Lineage , Cell Movement , Female , Forkhead Transcription Factors/genetics , Genetic Vectors , Male , Mice , Mice, Transgenic , Muscle Development/physiology , Muscle, Skeletal/metabolism , PAX3 Transcription Factor , Signal Transduction
17.
Hum Mol Genet ; 23(19): 5087-101, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24821700

ABSTRACT

Congenital heart defects affect at least 0.8% of newborn children and are a major cause of lethality prior to birth. Malformations of the arterial pole are particularly frequent. The myocardium at the base of the pulmonary trunk and aorta and the arterial tree associated with these great arteries are derived from splanchnic mesoderm of the second heart field (SHF), an important source of cardiac progenitor cells. These cells are controlled by a gene regulatory network that includes Fgf8, Fgf10 and Tbx1. Prdm1 encodes a transcriptional repressor that we show is also expressed in the SHF. In mouse embryos, mutation of Prdm1 affects branchial arch development and leads to persistent truncus arteriosus (PTA), indicative of neural crest dysfunction. Using conditional mutants, we show that this is not due to a direct function of Prdm1 in neural crest cells. Mutation of Prdm1 in the SHF does not result in PTA, but leads to arterial pole defects, characterized by mis-alignment or reduction of the aorta and pulmonary trunk, and abnormalities in the arterial tree, defects that are preceded by a reduction in outflow tract size and loss of caudal pharyngeal arch arteries. These defects are associated with a reduction in proliferation of progenitor cells in the SHF. We have investigated genetic interactions with Fgf8 and Tbx1, and show that on a Tbx1 heterozygote background, conditional Prdm1 mutants have more pronounced arterial pole defects, now including PTA. Our results identify PRDM1 as a potential modifier of phenotypic severity in TBX1 haploinsufficient DiGeorge syndrome patients.


Subject(s)
Epistasis, Genetic , Heart/embryology , Mesoderm/metabolism , Morphogenesis/genetics , T-Box Domain Proteins/genetics , Transcription Factors/genetics , Animals , Aorta, Thoracic/embryology , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Branchial Region/blood supply , Branchial Region/embryology , Branchial Region/metabolism , Branchial Region/pathology , Embryo, Mammalian , Female , Gene Expression , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Genotype , Male , Mice , Mice, Transgenic , Mutation , Organogenesis , Positive Regulatory Domain I-Binding Factor 1 , Stem Cells/metabolism , T-Box Domain Proteins/metabolism , Transcription Factors/metabolism
18.
PLoS One ; 9(2): e89397, 2014.
Article in English | MEDLINE | ID: mdl-24586749

ABSTRACT

The ecotropic viral integration site 1 (Evi1) oncogenic transcription factor is one of a number of alternative transcripts encoded by the Mds1 and Evi1 complex locus (Mecom). Overexpression of Evi1 has been observed in a number of myeloid disorders and is associated with poor patient survival. It is also amplified and/or overexpressed in many epithelial cancers including nasopharyngeal carcinoma, ovarian carcinoma, ependymomas, and lung and colorectal cancers. Two murine knockout models have also demonstrated Evi1's critical role in the maintenance of hematopoietic stem cell renewal with its absence resulting in the death of mutant embryos due to hematopoietic failure. Here we characterize a novel mouse model (designated Evi1(fl3)) in which Evi1 exon 3, which carries the ATG start, is flanked by loxP sites. Unexpectedly, we found that germline deletion of exon3 produces a hypomorphic allele due to the use of an alternative ATG start site located in exon 4, resulting in a minor Evi1 N-terminal truncation and a block in expression of the Mds1-Evi1 fusion transcript. Evi1(δex3/δex3) mutant embryos showed only a mild non-lethal hematopoietic phenotype and bone marrow failure was only observed in adult Vav-iCre/+, Evi1(fl3/fl3) mice in which exon 3 was specifically deleted in the hematopoietic system. Evi1(δex3/δex3) knockout pups are born in normal numbers but die during the perinatal period from congenital heart defects. Database searches identified 143 genes with similar mutant heart phenotypes as those observed in Evi1(δex3/δex3) mutant pups. Interestingly, 42 of these congenital heart defect genes contain known Evi1-binding sites, and expression of 18 of these genes are also effected by Evi1 siRNA knockdown. These results show a potential functional involvement of Evi1 target genes in heart development and indicate that Evi1 is part of a transcriptional program that regulates cardiac development in addition to the development of blood.


Subject(s)
Alleles , DNA-Binding Proteins/genetics , Genetic Association Studies , Heart Defects, Congenital/genetics , Proto-Oncogenes/genetics , Transcription Factors/genetics , Animals , Animals, Newborn , Base Sequence , Bone Marrow/pathology , DNA-Binding Proteins/chemistry , Disease Models, Animal , Exons , Gene Deletion , Gene Expression Regulation, Developmental , Genes, Lethal , Heart Defects, Congenital/mortality , Heart Defects, Congenital/pathology , Heart Defects, Congenital/physiopathology , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Immunophenotyping , MDS1 and EVI1 Complex Locus Protein , Mice , Mice, Knockout , Molecular Sequence Data , Mutation , Phenotype , Sequence Alignment , Severity of Illness Index , Transcription Factors/chemistry
19.
Mol Biol Evol ; 29(10): 3181-91, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22522309

ABSTRACT

Muscles are composed of multinucleated muscle fibers with different contractile and physiological properties, which result from specific slow or fast gene expression programs in the differentiated muscle cells. In the zebra fish embryo, the slow program is under the control of Hedgehog signaling from the notochord and floor plate. This pathway activates the expression of the conserved transcriptional repressor, Prdm1 (Blimp1), which in turn represses the fast program and promotes the slow program in adaxial cells of the somite and their descendants. In the mouse embryo, myogenesis is also initiated in the myotomal compartment of the somite, but the slow muscle program is not confined to a specific subset of cells. We now show that Prdm1 is expressed in the first differentiated myocytes of the early myotome from embryonic day (E)9.5-E11.5. During this period, muscle formation depends on the myogenic regulatory factors, Myf5 and Mrf4. In their absence, Prdm1 is not activated, in apparent contrast to zebra fish where Prdm1 is expressed in the absence of Myf5 and MyoD that drive myogenesis in adaxial cells. However, as in zebra fish, Prdm1 expression in the mouse myotome does not occur in the absence of Hedgehog signaling. Analysis of the muscle phenotype of Prdm1 mutant embryos shows that myogenesis appears to proceed normally. Notably, there is no requirement for Prdm1 activation of the slow muscle program in the mouse myotome. Furthermore, the gene for the transcriptional repressor, Sox6, which is repressed by Prdm1 to permit slow muscle differentiation in zebra fish, is not expressed in the mouse myotome. We propose that the lack of functional conservation for mouse Prdm1, that can nevertheless partially rescue the adaxial cells of zebra fish Prdm1 mutants, reflects differences in the evolution of the role of key regulators such as Prdm1 or Sox6, in initiating the onset of the slow muscle program, between teleosts and mammals.


Subject(s)
Conserved Sequence/genetics , Embryo, Mammalian/metabolism , Evolution, Molecular , Muscle Development/genetics , Muscle Fibers, Slow-Twitch/metabolism , Transcription Factors/genetics , Animals , Cell Differentiation/genetics , Embryo, Mammalian/cytology , Gene Expression Regulation, Developmental , Genes, Dominant/genetics , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Mice , Muscle Cells/cytology , Muscle Cells/metabolism , Muscle Fibers, Fast-Twitch/cytology , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Slow-Twitch/cytology , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Positive Regulatory Domain I-Binding Factor 1 , RNA, Messenger/genetics , RNA, Messenger/metabolism , SOXD Transcription Factors/genetics , SOXD Transcription Factors/metabolism , Signal Transduction/genetics , Transcription Factors/metabolism , Zebrafish/metabolism
20.
Curr Top Dev Biol ; 90: 1-41, 2010.
Article in English | MEDLINE | ID: mdl-20691846

ABSTRACT

The formation of the heart is a complex morphogenetic process that depends on the spatiotemporally regulated contribution of cardiac progenitor cells. These mainly derive from the splanchnic mesoderm of the first and second heart field (SHF), with an additional contribution of neurectodermally derived neural crest cells that are critical for the maturation of the arterial pole of the heart. The origin and distinguishing characteristics of the two heart fields, as well as the relation of the SHF to the proepicardial organ and to a proposed third heart field are still subjects of debate. In the last ten years many genes that function in the SHF have been identified, leading to the establishment of a gene regulatory network in the mouse embryo. It is becoming increasingly evident that distinct gene networks control subdomains of the SHF that contribute to different parts of the heart. Although there is now extensive information about mutant phenotypes that reflect problems in the integration of progenitor cells into the developing heart, relatively little is known about the mechanisms that regulate SHF cell behavior. This important source of cardiac progenitor cells must be maintained as a proliferative, undifferentiated cell population. Selected subpopulations, at different development stages, are directed to myocardial, and also to smooth muscle and endothelial cell fates, as they integrate into the heart. Analysis of signaling pathways that impact the SHF, as well as regulatory factors, is beginning to reveal mechanisms that control cardiac progenitor cell behavior.


Subject(s)
Heart/embryology , Myocardium/cytology , Stem Cells/physiology , Animals , Biomarkers/metabolism , Cell Proliferation , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Gene Expression Regulation, Developmental , Heart/anatomy & histology , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Morphogenesis/physiology , Neural Crest/cytology , Neural Crest/metabolism , Signal Transduction/physiology , Stem Cells/cytology , Wnt Proteins/genetics , Wnt Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...