Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Development ; 149(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-35043944

ABSTRACT

Establishment of a healthy ovarian reserve is contingent upon numerous regulatory pathways during embryogenesis. Previously, mice lacking TBP-associated factor 4b (Taf4b) were shown to exhibit a diminished ovarian reserve. However, potential oocyte-intrinsic functions of TAF4b have not been examined. Here, we use a combination of gene expression profiling and chromatin mapping to characterize TAF4b-dependent gene regulatory networks in mouse oocytes. We find that Taf4b-deficient oocytes display inappropriate expression of meiotic, chromatin modification/organization, and X-linked genes. Furthermore, dysregulated genes in Taf4b-deficient oocytes exhibit an unexpected amount of overlap with dysregulated genes in oocytes from XO female mice, a mouse model of Turner Syndrome. Using Cleavage Under Targets and Release Using Nuclease (CUT&RUN), we observed TAF4b enrichment at genes involved in chromatin remodeling and DNA repair, some of which are differentially expressed in Taf4b-deficient oocytes. Interestingly, TAF4b target genes were enriched for Sp/Klf family and NFY target motifs rather than TATA-box motifs, suggesting an alternative mode of promoter interaction. Together, our data connect several gene regulatory nodes that contribute to the precise development of the mammalian ovarian reserve.


Subject(s)
Gene Regulatory Networks/genetics , Oogenesis , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics , Animals , DNA Repair , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Germ Cells/cytology , Germ Cells/metabolism , Meiosis , Mice , Mice, Inbred C57BL , Mice, Knockout , Oocytes/cytology , Oocytes/metabolism , Promoter Regions, Genetic , TATA-Binding Protein Associated Factors/deficiency , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/deficiency , Transcription Factor TFIID/metabolism , X Chromosome/genetics , X Chromosome/metabolism
2.
Mol Cell ; 68(1): 118-129.e5, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28918900

ABSTRACT

Previous studies suggested that expression of most yeast mRNAs is dominated by either transcription factor TFIID or SAGA. We re-examined the role of TFIID by rapid depletion of S. cerevisiae TFIID subunits and measurement of changes in nascent transcription. We find that transcription of nearly all mRNAs is strongly dependent on TFIID function. Degron-dependent depletion of Taf1, Taf2, Taf7, Taf11, and Taf13 showed similar transcription decreases for genes in the Taf1-depleted, Taf1-enriched, TATA-containing, and TATA-less gene classes. The magnitude of TFIID dependence varies with growth conditions, although this variation is similar genome-wide. Many studies have suggested differences in gene-regulatory mechanisms between TATA and TATA-less genes, and these differences have been attributed in part to differential dependence on SAGA or TFIID. Our work indicates that TFIID participates in expression of nearly all yeast mRNAs and that differences in regulation between these two gene categories is due to other properties.


Subject(s)
Gene Expression Regulation, Fungal , Genome, Fungal , RNA Polymerase II/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , TATA-Box Binding Protein/genetics , Trans-Activators/chemistry , Transcription, Genetic , Gene Deletion , Promoter Regions, Genetic , Protein Subunits/genetics , Protein Subunits/metabolism , RNA Polymerase II/metabolism , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , TATA-Binding Protein Associated Factors/deficiency , TATA-Binding Protein Associated Factors/genetics , TATA-Box Binding Protein/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factor TFIID/deficiency , Transcription Factor TFIID/genetics
3.
Neurobiol Dis ; 149: 105224, 2021 02.
Article in English | MEDLINE | ID: mdl-33359140

ABSTRACT

The TATA-box binding protein associated factor 1 (TAF1) is part of the TFIID complex that plays a key role during the initiation of transcription. Variants of TAF1 are associated with neurodevelopmental disorders. Previously, we found that CRISPR/Cas9 based editing of the TAF1 gene disrupts the morphology of the cerebral cortex and blunts the expression as well as the function of the CaV3.1 (T-type) voltage gated calcium channel. Here, we tested the efficacy of SAK3 (ethyl 8'-methyl-2', 4-dioxo-2-(piperidin-1-yl)-2'H-spiro [cyclopentane-1, 3'-imidazo [1, 2-a] pyridine]-2-ene-3-carboxylate), a T-type calcium channel enhancer, in an animal model of TAF1 intellectual disability (ID) syndrome. At post-natal day 3, rat pups were subjected to intracerebroventricular (ICV) injection of either gRNA-control or gRNA-TAF1 CRISPR/Cas9 viruses. At post-natal day 21, the rat pups were given SAK3 (0.25 mg/kg, p.o.) or vehicle for 14 days (i.e. till post-natal day 35) and then subjected to behavioral, morphological, and molecular studies. Oral administration of SAK3 (0.25 mg/kg, p.o.) significantly rescued locomotion abnormalities associated with TAF1 gene editing. SAK3 treatment prevented the loss of cortical neurons and GFAP-positive astrocytes observed after TAF1 gene editing. In addition, SAK3 protected cells from apoptosis. SAK3 also restored the Brain-derived neurotrophic factor/protein kinase B/Glycogen Synthase Kinase 3 Beta (BDNF/AKT/GSK3ß) signaling axis in TAF1 edited animals. Finally, SAK3 normalized the levels of three GSK3ß substrates - CaV3.1, FOXP2, and CRMP2. We conclude that the T-type calcium channel enhancer SAK3 is beneficial against the deleterious effects of TAF1 gene-editing, in part, by stimulating the BDNF/AKT/GSK3ß signaling pathway.


Subject(s)
Calcium Channels, T-Type/metabolism , Disease Models, Animal , Histone Acetyltransferases/deficiency , Imidazoles/administration & dosage , Intellectual Disability/drug therapy , Intellectual Disability/metabolism , Spiro Compounds/administration & dosage , TATA-Binding Protein Associated Factors/deficiency , Transcription Factor TFIID/deficiency , Animals , Animals, Newborn , Drug Evaluation, Preclinical/methods , Female , Histone Acetyltransferases/genetics , Injections, Intraventricular , Intellectual Disability/genetics , Locomotion/drug effects , Locomotion/physiology , Pregnancy , Rats , Rats, Sprague-Dawley , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics
4.
Exp Cell Res ; 371(2): 311-321, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30092221

ABSTRACT

Mutations in the Fused in Sarcoma (FUS) gene have been identified in familial ALS in human. Drosophila contains a single ortholog of human FUS called Cabeza (Caz). We previously established Drosophila models of ALS targeted to Caz, which developed the locomotive dysfunction and caused anatomical defects in presynaptic terminals of motoneurons. Accumulating evidence suggests that ALS and cancer share defects in many cellular processes. The Hippo pathway was originally discovered in Drosophila and plays a role as a tumor suppressor in mammals. We aimed to determine whether Hippo pathway genes modify the ALS phenotype using Caz knockdown flies. We found a genetic link between Caz and Hippo (hpo), the Drosophila ortholog of human Mammalian sterile 20-like kinase (MST) 1 and 2. Loss-of-function mutations of hpo rescued Caz knockdown-induced eye- and neuron-specific defects. The decreased Caz levels in nuclei induced by Caz knockdown were also rescued by loss of function mutations of hpo. Moreover, hpo mRNA level was dramatically increased in Caz knockdown larvae, indicating that Caz negatively regulated hpo. Our results demonstrate that hpo, Drosophila MST, is a novel modifier of Drosophila FUS. Therapeutic targets that inhibit the function of MST could modify the pathogenic processes of ALS.


Subject(s)
Cell Cycle Proteins/genetics , Cytoskeletal Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Intracellular Signaling Peptides and Proteins/genetics , Larva/genetics , Neurogenesis/genetics , Protein Serine-Threonine Kinases/genetics , RNA-Binding Proteins/genetics , Transcription Factor TFIID/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Cycle Proteins/metabolism , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Drosophila Proteins/deficiency , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Eye/metabolism , Eye/ultrastructure , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Larva/cytology , Larva/growth & development , Larva/metabolism , Motor Neurons/metabolism , Motor Neurons/pathology , Nerve Degeneration , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Transcription Factor TFIID/deficiency
5.
Nature ; 495(7442): 516-9, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23503660

ABSTRACT

Embryonic stem (ES) cells are pluripotent and characterized by open chromatin and high transcription levels, achieved through auto-regulatory and feed-forward transcription factor loops. ES-cell identity is maintained by a core of factors including Oct4 (also known as Pou5f1), Sox2, Klf4, c-Myc (OSKM) and Nanog, and forced expression of the OSKM factors can reprogram somatic cells into induced pluripotent stem cells (iPSCs) resembling ES cells. These gene-specific factors for RNA-polymerase-II-mediated transcription recruit transcriptional cofactors and chromatin regulators that control access to and activity of the basal transcription machinery on gene promoters. How the basal transcription machinery is involved in setting and maintaining the pluripotent state is unclear. Here we show that knockdown of the transcription factor IID (TFIID) complex affects the pluripotent circuitry in mouse ES cells and inhibits reprogramming of fibroblasts. TFIID subunits and the OSKM factors form a feed-forward loop to induce and maintain a stable transcription state. Notably, transient expression of TFIID subunits greatly enhanced reprogramming. These results show that TFIID is critical for transcription-factor-mediated reprogramming. We anticipate that, by creating plasticity in gene expression programs, transcription complexes such as TFIID assist reprogramming into different cellular states.


Subject(s)
Pluripotent Stem Cells/metabolism , Transcription Factor TFIID/metabolism , Transcription, Genetic , Animals , Cellular Reprogramming/genetics , Chromatin/genetics , Chromatin/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Male , Mice , Pluripotent Stem Cells/cytology , Promoter Regions, Genetic/genetics , RNA Polymerase II/metabolism , TATA-Binding Protein Associated Factors/deficiency , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism , TATA-Box Binding Protein/metabolism , Transcription Factor TFIID/deficiency , Transcription Factor TFIID/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Dis Model Mech ; 17(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38804708

ABSTRACT

The TATA box-binding protein-associated factor 1 (TAF1) is a ubiquitously expressed protein and the largest subunit of the basal transcription factor TFIID, which plays a key role in initiation of RNA polymerase II-dependent transcription. TAF1 missense variants in human males cause X-linked intellectual disability, a neurodevelopmental disorder, and TAF1 is dysregulated in X-linked dystonia-parkinsonism, a neurodegenerative disorder. However, this field has lacked a genetic mouse model of TAF1 disease to explore its mechanism in mammals and treatments. Here, we generated and validated a conditional cre-lox allele and the first ubiquitous Taf1 knockout mouse. We discovered that Taf1 deletion in male mice was embryonically lethal, which may explain why no null variants have been identified in humans. In the brains of Taf1 heterozygous female mice, no differences were found in gross structure, overall expression and protein localisation, suggesting extreme skewed X inactivation towards the non-mutant chromosome. Nevertheless, these female mice exhibited a significant increase in weight, weight with age, and reduced movement, suggesting that a small subset of neurons was negatively impacted by Taf1 loss. Finally, this new mouse model may be a future platform for the development of TAF1 disease therapeutics.


Subject(s)
Body Weight , Heterozygote , Histone Acetyltransferases , Mice, Knockout , Movement Disorders , TATA-Binding Protein Associated Factors , Transcription Factor TFIID , Animals , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism , Transcription Factor TFIID/deficiency , Female , Male , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Movement Disorders/genetics , Movement Disorders/pathology , Embryo, Mammalian/metabolism , Mice , Brain/pathology , Brain/metabolism , Genes, Lethal , Mice, Inbred C57BL
7.
Cell Death Dis ; 12(8): 790, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34385420

ABSTRACT

Regulation of gene expression involves a complex and dynamic dialogue between transcription factors, chromatin remodelling and modification complexes and the basal transcription machinery. To address the function of the Taf4 subunit of general transcription factor TFIID in the regulation of insulin signalling, it was inactivated in adult murine pancreatic beta cells. Taf4 inactivation impacted the expression of critical genes involved in beta-cell function leading to increased glycaemia, lowered plasma insulin levels and defective glucose-stimulated insulin secretion. One week after Taf4-loss, single-cell RNA-seq revealed cells with mixed beta cell, alpha and/or delta cell identities as well as a beta cell population trans-differentiating into alpha-like cells. Computational analysis of single-cell RNA-seq defines how known critical beta cell and alpha cell determinants may act in combination with additional transcription factors and the NuRF chromatin remodelling complex to promote beta cell trans-differentiation.


Subject(s)
Cell Transdifferentiation/genetics , Gene Expression Profiling , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Protein Subunits/metabolism , Single-Cell Analysis , Transcription Factor TFIID/metabolism , Aging/metabolism , Animals , Body Weight/drug effects , Cell Transdifferentiation/drug effects , Gene Expression Regulation/drug effects , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/metabolism , Glucose/pharmacology , Insulin/metabolism , Insulin Secretion/drug effects , Insulin-Secreting Cells/drug effects , Mice , Mutation/genetics , Transcription Factor TFIID/deficiency , Transcription Factors/metabolism
8.
Nat Commun ; 7: 11063, 2016 Mar 30.
Article in English | MEDLINE | ID: mdl-27026076

ABSTRACT

TAF4 (TATA-binding protein-associated factor 4) and its paralogue TAF4b are components of the TFIID core module. We inactivated the murine Taf4a gene to address Taf4 function during embryogenesis. Here we show that Taf4a(-/-) embryos survive until E9.5 where primary germ layers and many embryonic structures are identified showing Taf4 is dispensable for their specification. In contrast, Taf4 is required for correct patterning of the trunk and anterior structures, ventral morphogenesis and proper heart positioning. Overlapping expression of Taf4a and Taf4b during embryogenesis suggests their redundancy at early stages. In agreement with this, Taf4a(-/-) embryonic stem cells (ESCs) are viable and comprise Taf4b-containing TFIID. Nevertheless, Taf4a(-/-) ESCs do not complete differentiation into glutamatergic neurons and cardiomyocytes in vitro due to impaired preinitiation complex formation at the promoters of critical differentiation genes. We define an essential role of a core TFIID TAF in differentiation events during mammalian embryogenesis.


Subject(s)
Cell Differentiation , Embryonic Development , Mouse Embryonic Stem Cells/metabolism , Protein Subunits/metabolism , Transcription Factor TFIID/metabolism , Animals , Biomarkers/metabolism , Body Patterning/drug effects , Body Patterning/genetics , Cardiovascular Abnormalities/embryology , Cardiovascular Abnormalities/genetics , Cardiovascular Abnormalities/pathology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Survival/drug effects , Embryo Loss/genetics , Embryo Loss/pathology , Embryonic Development/drug effects , Embryonic Development/genetics , Female , Gene Expression Regulation, Developmental/drug effects , Germ Cells/drug effects , Germ Cells/metabolism , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/drug effects , Mutation , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Neural Crest/drug effects , Neural Crest/pathology , Neurogenesis/drug effects , Neurogenesis/genetics , Neurons/drug effects , Neurons/metabolism , Phenotype , Pregnancy , Protein Subunits/genetics , Transcription Factor TFIID/deficiency , Transcription Factor TFIID/genetics , Tretinoin/pharmacology
9.
Elife ; 3: e03613, 2014 Sep 10.
Article in English | MEDLINE | ID: mdl-25209997

ABSTRACT

The functions of the TAF subunits of mammalian TFIID in physiological processes remain poorly characterised. In this study, we describe a novel function of TAFs in directing genomic occupancy of a transcriptional activator. Using liver-specific inactivation in mice, we show that the TAF4 subunit of TFIID is required for post-natal hepatocyte maturation. TAF4 promotes pre-initiation complex (PIC) formation at post-natal expressed liver function genes and down-regulates a subset of embryonic expressed genes by increased RNA polymerase II pausing. The TAF4-TAF12 heterodimer interacts directly with HNF4A and in vivo TAF4 is necessary to maintain HNF4A-directed embryonic gene expression at post-natal stages and promotes HNF4A occupancy of functional cis-regulatory elements adjacent to the transcription start sites of post-natal expressed genes. Stable HNF4A occupancy of these regulatory elements requires TAF4-dependent PIC formation highlighting that these are mutually dependent events. Local promoter-proximal HNF4A-TFIID interactions therefore act as instructive signals for post-natal hepatocyte differentiation.


Subject(s)
Cell Differentiation/genetics , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Promoter Regions, Genetic , Protein Subunits/metabolism , Transcription Factor TFIID/metabolism , Animals , Animals, Newborn , Bile Ducts/metabolism , Bile Ducts/pathology , Cell Communication , Cholestasis/complications , Cholestasis/metabolism , Cholestasis/pathology , Down-Regulation/genetics , Genome , Hepatocyte Nuclear Factor 4/chemistry , Homeodomain Proteins/metabolism , Hypoglycemia/complications , Hypoglycemia/pathology , Mice , Mutation/genetics , Protein Binding/genetics , Protein Multimerization , Protein Structure, Tertiary , RNA Polymerase II/metabolism , TATA-Binding Protein Associated Factors , TATA-Box Binding Protein/metabolism , Transcription Factor TFIID/deficiency , Up-Regulation/genetics
10.
Elife ; 2: e00170, 2013 Jan 08.
Article in English | MEDLINE | ID: mdl-23326641

ABSTRACT

The diverse transcriptional mechanisms governing cellular differentiation and development of mammalian tissue remains poorly understood. Here we report that TAF7L, a paralogue of TFIID subunit TAF7, is enriched in adipocytes and white fat tissue (WAT) in mouse. Depletion of TAF7L reduced adipocyte-specific gene expression, compromised adipocyte differentiation, and WAT development as well. Ectopic expression of TAF7L in myoblasts reprograms these muscle precursors into adipocytes upon induction. Genome-wide mRNA-seq expression profiling and ChIP-seq binding studies confirmed that TAF7L is required for activating adipocyte-specific genes via a dual mechanism wherein it interacts with PPARγ at enhancers and TBP/Pol II at core promoters. In vitro binding studies confirmed that TAF7L forms complexes with both TBP and PPARγ. These findings suggest that TAF7L plays an integral role in adipocyte gene expression by targeting enhancers as a cofactor for PPARγ and promoters as a component of the core transcriptional machinery.DOI:http://dx.doi.org/10.7554/eLife.00170.001.


Subject(s)
Adipocytes/metabolism , Adipogenesis , Adipose Tissue, White/metabolism , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Transcription Factors/metabolism , 3T3-L1 Cells , Adipogenesis/genetics , Adipose Tissue, White/cytology , Animals , Binding Sites , Cellular Reprogramming , Chromatin Immunoprecipitation , Enhancer Elements, Genetic , Gene Expression Profiling , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Myoblasts/metabolism , PPAR gamma/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , Signal Transduction , TATA-Binding Protein Associated Factors/deficiency , TATA-Binding Protein Associated Factors/genetics , TATA-Box Binding Protein/metabolism , Transcription Factor TFIID/deficiency , Transcription Factor TFIID/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic , Transfection
11.
Dev Biol ; 303(2): 715-26, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17207475

ABSTRACT

Oocyte development in the mammalian ovary requires productive interactions with somatic granulosa cells of the ovarian follicle. Proliferating granulosa cells support the progression of follicular growth and maturation, multiplying dramatically as it unfolds. The cell cycle recruitment of granulosa cells is regulated at least in part by hormones such as follicle-stimulating hormone (FSH) and estrogen. Follicles recruited into the growth phase following formation of multiple layers of granulosa cells have two major fates: either to continue proliferation followed by differentiation, or to die by programmed cell death, or atresia. While many of the signaling pathways orchestrating ovarian follicle development are known, the downstream transcriptional regulators that integrate such signals in the mammalian ovary remain to be defined. Recent experiments in diverse organisms have revealed multiple instances of gonad-selective components of the basal transcriptional machinery. One such protein, TAF4b, is a gonadal-enriched coactivator subunit of the TFIID complex required for normal female fertility in the mouse. To determine the etiology of female infertility of the TAF4b-deficient mice, we have determined multiple functions of TAF4b during postnatal ovarian follicle development. Here we demonstrate that the TAF4b protein is expressed in the granulosa cell compartment of the mammalian ovarian follicle. Furthermore, TAF4b-deficient mouse ovaries contain reduced numbers of primordial as well as growing follicles and a concomitant increased proportion of apoptotic follicles in comparison to wild type counterparts. Importantly, TAF4b-null follicles are largely resistant to induction of proliferation in response to multiple hormonal stimuli including estrogen and FSH and demonstrate compromised granulosa cell survival. Together, these data suggest that TAF4b integrates a program of granulosa cell gene expression required for normal ovarian follicle survival and proliferation in response to diverse ovarian signaling events.


Subject(s)
Granulosa Cells/cytology , Granulosa Cells/metabolism , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Animals , Base Sequence , Cell Proliferation , Cell Survival/genetics , Cell Survival/physiology , DNA Primers/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovarian Follicle/cytology , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Ovary/cytology , Ovary/growth & development , Ovary/metabolism , Signal Transduction , TATA-Binding Protein Associated Factors/deficiency , TATA-Binding Protein Associated Factors/genetics , Tissue Distribution , Transcription Factor TFIID/deficiency , Transcription Factor TFIID/genetics
12.
Dev Biol ; 285(1): 28-37, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16039642

ABSTRACT

TFIID, composed of the TATA box binding protein (TBP) and 13 TBP-associated factors (TAFs), plays a role in nucleating the assembly of the RNA polymerase II preinitiation complexes on protein coding genes. TAF10 (formerly TAF(II)30) is shared between TFIID and other transcription regulatory complexes (i.e. SAGA, TFTC, STAGA and PCAF/GCN5). TAF10 is an essential transcription factor during very early stages of mouse embryo development. To study the in vivo function of TAF10 in cellular differentiation and proliferation at later stages, the role of TAF10 was analysed in keratinocytes during skin development and adult epidermal homeostasis. We demonstrate that ablation of TAF10 in keratinocytes of the forming epidermis affects the expression of some, but not all genes, impairs keratinocyte terminal differentiation and alters skin permeability barrier functions. In contrast, loss of TAF10 in keratinocytes of adult epidermis did not (i) modify the expression of tested genes, (ii) affect epidermal homeostasis and (iii) impair acute response to UV irradiation or skin regeneration after wounding. Thus, this study demonstrates for the first time a differential in vivo requirement for a mammalian TAF for the regulation of gene expression depending on the cellular environment and developmental stage of the cell.


Subject(s)
Skin Physiological Phenomena , TATA-Binding Protein Associated Factors/physiology , Transcription Factor TFIID/physiology , Animals , Animals, Newborn , Base Sequence , Cell Differentiation , Cell Proliferation , DNA, Complementary/genetics , Female , Fetus/cytology , Gene Expression Regulation, Developmental , Keratinocytes/cytology , Male , Mice , Mice, Knockout , Mice, Transgenic , Permeability , Phenotype , Pregnancy , Skin/cytology , Skin/embryology , Skin/growth & development , TATA-Binding Protein Associated Factors/deficiency , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/deficiency , Transcription Factor TFIID/genetics
13.
Genes Dev ; 19(7): 794-803, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15774719

ABSTRACT

The establishment and maintenance of spermatogenesis in mammals requires specialized networks of gene expression programs in the testis. The gonad-specific TAF4b component of TFIID (formerly TAF(II)105) is a transcriptional regulator enriched in the mouse testis. Herein we show that TAF4b is required for maintenance of spermatogenesis in the mouse. While young Taf4b-null males are initially fertile, Taf4b-null males become infertile by 3 mo of age and eventually exhibit seminiferous tubules devoid of germ cells. At birth, testes of Taf4b-null males appear histologically normal; however, at post-natal day 3 gonocyte proliferation is impaired and expression of spermatogonial stem cell markers c-Ret, Plzf, and Stra8 is reduced. Together, these data indicate that TAF4b is required for the precise expression of gene products essential for germ cell proliferation and suggest that TAF4b may be required for the regulation of spermatogonial stem cell specification and proliferation that is obligatory for normal spermatogenic maintenance in the adult.


Subject(s)
Infertility, Male/genetics , Spermatogenesis/physiology , TATA-Binding Protein Associated Factors/metabolism , Testis/metabolism , Transcription Factor TFIID/metabolism , Aging/pathology , Animals , Cell Division/physiology , Male , Mice , Spermatids/metabolism , Spermatogenesis/genetics , Spermatogonia/cytology , Spermatogonia/metabolism , TATA-Binding Protein Associated Factors/deficiency , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/deficiency , Transcription Factor TFIID/genetics
SELECTION OF CITATIONS
SEARCH DETAIL