Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Stem Cells ; 38(2): 202-217, 2020 02.
Article in English | MEDLINE | ID: mdl-31675135

ABSTRACT

Cooperative actions of extrinsic signals and cell-intrinsic transcription factors alter gene regulatory networks enabling cells to respond appropriately to environmental cues. Signaling by transforming growth factor type ß (TGFß) family ligands (eg, bone morphogenetic proteins [BMPs] and Activin/Nodal) exerts cell-type specific and context-dependent transcriptional changes, thereby steering cellular transitions throughout embryogenesis. Little is known about coordinated regulation and transcriptional interplay of the TGFß system. To understand intrafamily transcriptional regulation as part of this system's actions during development, we selected 95 of its components and investigated their mRNA-expression dynamics, gene-gene interactions, and single-cell expression heterogeneity in mouse embryonic stem cells transiting to neural progenitors. Interrogation at 24 hour intervals identified four types of temporal gene transcription profiles that capture all stages, that is, pluripotency, epiblast formation, and neural commitment. Then, between each stage we performed esiRNA-based perturbation of each individual component and documented the effect on steady-state mRNA levels of the remaining 94 components. This exposed an intricate system of multilevel regulation whereby the majority of gene-gene interactions display a marked cell-stage specific behavior. Furthermore, single-cell RNA-profiling at individual stages demonstrated the presence of detailed co-expression modules and subpopulations showing stable co-expression modules such as that of the core pluripotency genes at all stages. Our combinatorial experimental approach demonstrates how intrinsically complex transcriptional regulation within a given pathway is during cell fate/state transitions.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Embryonic Stem Cells/metabolism , Transforming Growth Factor beta/metabolism , Cell Differentiation , Humans
2.
Brain ; 142(9): 2631-2643, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31334757

ABSTRACT

Marked by incomplete division of the embryonic forebrain, holoprosencephaly is one of the most common human developmental disorders. Despite decades of phenotype-driven research, 80-90% of aneuploidy-negative holoprosencephaly individuals with a probable genetic aetiology do not have a genetic diagnosis. Here we report holoprosencephaly associated with variants in the two X-linked cohesin complex genes, STAG2 and SMC1A, with loss-of-function variants in 10 individuals and a missense variant in one. Additionally, we report four individuals with variants in the cohesin complex genes that are not X-linked, SMC3 and RAD21. Using whole mount in situ hybridization, we show that STAG2 and SMC1A are expressed in the prosencephalic neural folds during primary neurulation in the mouse, consistent with forebrain morphogenesis and holoprosencephaly pathogenesis. Finally, we found that shRNA knockdown of STAG2 and SMC1A causes aberrant expression of HPE-associated genes ZIC2, GLI2, SMAD3 and FGFR1 in human neural stem cells. These findings show the cohesin complex as an important regulator of median forebrain development and X-linked inheritance patterns in holoprosencephaly.


Subject(s)
Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Holoprosencephaly/diagnosis , Holoprosencephaly/genetics , Adolescent , Animals , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Mice , Mice, Inbred C57BL , Cohesins
3.
J Biol Chem ; 293(8): 2711-2724, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29284678

ABSTRACT

Chromatin in embryonic stem cells (ESCs) differs markedly from that in somatic cells, with ESCs exhibiting a more open chromatin configuration. Accordingly, ATP-dependent chromatin remodeling complexes are important regulators of ESC homeostasis. Depletion of the remodeler SMARCAD1, an ATPase of the SNF2 family, has been shown to affect stem cell state, but the mechanistic explanation for this effect is unknown. Here, we set out to gain further insights into the function of SMARCAD1 in mouse ESCs. We identified KRAB-associated protein 1 (KAP1) as the stoichiometric binding partner of SMARCAD1 in ESCs. We found that this interaction occurs on chromatin and that SMARCAD1 binds to different classes of KAP1 target genes, including zinc finger protein (ZFP) and imprinted genes. We also found that the RING B-box coiled-coil (RBCC) domain in KAP1 and the proximal coupling of ubiquitin conjugation to ER degradation (CUE) domain in SMARCAD1 mediate their direct interaction. Of note, retention of SMARCAD1 in the nucleus depended on KAP1 in both mouse ESCs and human somatic cells. Mutations in the CUE1 domain of SMARCAD1 perturbed the binding to KAP1 in vitro and in vivo Accordingly, an intact CUE1 domain was required for tethering this remodeler to the nucleus. Moreover, mutation of the CUE1 domain compromised SMARCAD1 binding to KAP1 target genes. Taken together, our results reveal a mechanism that localizes SMARCAD1 to genomic sites through the interaction of SMARCAD1's CUE1 motif with KAP1.


Subject(s)
Adult Stem Cells/metabolism , Cell Nucleus/metabolism , DNA Helicases/metabolism , Gene Expression Regulation , Mouse Embryonic Stem Cells/metabolism , Tripartite Motif-Containing Protein 28/metabolism , Adult Stem Cells/cytology , Adult Stem Cells/enzymology , Amino Acid Substitution , Animals , Cell Line , Cell Nucleus/enzymology , Chromatin/chemistry , Chromatin/enzymology , Chromatin/metabolism , Chromatin Assembly and Disassembly , DNA Helicases/antagonists & inhibitors , DNA Helicases/chemistry , DNA Helicases/genetics , Gene Deletion , Humans , Kinetics , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/enzymology , Mutation , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Tripartite Motif-Containing Protein 28/antagonists & inhibitors , Tripartite Motif-Containing Protein 28/chemistry , Tripartite Motif-Containing Protein 28/genetics
5.
Cell Rep ; 26(7): 1906-1918.e8, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30759399

ABSTRACT

In this study, we demonstrate that, among all five CBX Polycomb proteins, only CBX7 possesses the ability to control self-renewal of human hematopoietic stem and progenitor cells (HSPCs). Xenotransplantation of CBX7-overexpressing HSPCs resulted in increased multi-lineage long-term engraftment and myelopoiesis. Gene expression and chromatin analyses revealed perturbations in genes involved in differentiation, DNA and chromatin maintenance, and cell cycle control. CBX7 is upregulated in acute myeloid leukemia (AML), and its genetic or pharmacological repression in AML cells inhibited proliferation and induced differentiation. Mass spectrometry analysis revealed several non-histone protein interactions between CBX7 and the H3K9 methyltransferases SETDB1, EHMT1, and EHMT2. These CBX7-binding proteins possess a trimethylated lysine peptide motif highly similar to the canonical CBX7 target H3K27me3. Depletion of SETDB1 in AML cells phenocopied repression of CBX7. We identify CBX7 as an important regulator of self-renewal and uncover non-canonical crosstalk between distinct pathways, revealing therapeutic opportunities for leukemia.


Subject(s)
Hematopoietic Stem Cells/metabolism , Polycomb Repressive Complex 1/metabolism , Stem Cells/metabolism , Animals , Female , Fetal Blood/cytology , Fetal Blood/metabolism , HEK293 Cells , HL-60 Cells , Hematopoietic Stem Cells/cytology , Heterografts , Histone-Lysine N-Methyltransferase/metabolism , Humans , K562 Cells , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Polycomb Repressive Complex 1/biosynthesis , Polycomb Repressive Complex 1/genetics , Stem Cells/cytology , Transcription, Genetic
6.
Nat Commun ; 10(1): 2669, 2019 06 17.
Article in English | MEDLINE | ID: mdl-31209209

ABSTRACT

The Mediator complex regulates transcription by connecting enhancers to promoters. High Mediator binding density defines super enhancers, which regulate cell-identity genes and oncogenes. Protein interactions of Mediator may explain its role in these processes but have not been identified comprehensively. Here, we purify Mediator from neural stem cells (NSCs) and identify 75 protein-protein interaction partners. We identify super enhancers in NSCs and show that Mediator-interacting chromatin modifiers colocalize with Mediator at enhancers and super enhancers. Transcription factor families with high affinity for Mediator dominate enhancers and super enhancers and can explain genome-wide Mediator localization. We identify E-box transcription factor Tcf4 as a key regulator of NSCs. Tcf4 interacts with Mediator, colocalizes with Mediator at super enhancers and regulates neurogenic transcription factor genes with super enhancers and broad H3K4me3 domains. Our data suggest that high binding-affinity for Mediator is an important organizing feature in the transcriptional network that determines NSC identity.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Gene Regulatory Networks/physiology , Mediator Complex/metabolism , Neural Stem Cells/physiology , Neurogenesis/genetics , Transcription Factor 4/metabolism , Cell Line , Enhancer Elements, Genetic/genetics , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Oxidoreductases, N-Demethylating/metabolism , Promoter Regions, Genetic/genetics , Protein Interaction Mapping , Protein Interaction Maps/genetics , Protein-Arginine N-Methyltransferases/metabolism , Transcription, Genetic/physiology
SELECTION OF CITATIONS
SEARCH DETAIL