Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep ; 43(4): 114083, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38602877

ABSTRACT

A common cause of deafness in humans is dysregulation of the endocochlear potential generated by the stria vascularis (SV). Thus, proper formation of the SV is critical for hearing. Using single-cell transcriptomics and a series of Shh signaling mutants, we discovered that the Shh receptor Patched1 (Ptch1) is essential for marginal cell (MC) differentiation and SV formation. Single-cell RNA sequencing analyses revealed that the cochlear roof epithelium is already specified into discrete domains with distinctive gene expression profiles at embryonic day 14, with Gsc as a marker gene of the MC lineage. Ptch1 deficiency leads to defective specification of MC precursors along the cochlear basal-apical regions. We demonstrated that elevated Gli2 levels impede MC differentiation through sustaining Otx2 expression and maintaining the progenitor state of MC precursors. Our results uncover an early specification of cochlear non-sensory epithelial cells and establish a crucial role of the Ptch1-Gli2 axis in regulating the development of SV.


Subject(s)
Cell Differentiation , Cochlea , Patched-1 Receptor , Stria Vascularis , Patched-1 Receptor/metabolism , Patched-1 Receptor/genetics , Animals , Mice , Stria Vascularis/metabolism , Stria Vascularis/cytology , Cochlea/metabolism , Cochlea/embryology , Cochlea/cytology , Signal Transduction , Zinc Finger Protein Gli2/metabolism , Zinc Finger Protein Gli2/genetics , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics
2.
Nat Commun ; 14(1): 6699, 2023 10 23.
Article in English | MEDLINE | ID: mdl-37872167

ABSTRACT

Increasing evidence has demonstrated that drug resistance can be acquired in cancer cells by kinase rewiring, which is an obstacle for efficient cancer therapy. However, it is technically challenging to measure the expression of protein kinases on large scale due to their dynamic range in human proteome. We employ a lysine-targeted sulfonyl fluoride probe, named XO44, which binds to 133 endogenous kinases in intact lenvatinib-resistant hepatocellular carcinoma (HCC) cells. This analysis reveals cyclin-dependent kinase 6 (CDK6) upregulation, which is mediated by ERK/YAP1 signaling cascade. Functional analyses show that CDK6 is crucial in regulation of acquired lenvatinib resistance in HCC via augmentation of liver cancer stem cells with clinical significance. We identify a noncanonical pathway of CDK6 in which it binds and regulates the activity of GSK3ß, leading to activation of Wnt/ß-catenin signaling. Consistently, CDK6 inhibition by palbociclib or degradation by proteolysis targeting chimeras (PROTACs) is highly synergistic with lenvatinib in vitro. Interestingly, palbociclib not only exerts maximal growth suppressive effect with lenvatinib in lenvatinib-resistant HCC models but also reshapes the tumor immune microenvironment. Together, we unveil CDK6 as a druggable target in lenvatinib-resistant HCC and highlight the use of a chemical biology approach to understand nongenetic resistance mechanisms in cancer.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Up-Regulation , Cyclin-Dependent Kinase 6/metabolism , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Cell Line, Tumor , Tumor Microenvironment
3.
Aging Cell ; 21(8): e13673, 2022 08.
Article in English | MEDLINE | ID: mdl-35851988

ABSTRACT

Muscle satellite cells (SCs) are responsible for muscle homeostasis and regeneration and lncRNAs play important roles in regulating SC activities. Here, in this study, we identify PAM (Pax7 Associated Muscle lncRNA) that is induced in activated/proliferating SCs upon injury to promote SC proliferation as myoblast cells. PAM is generated from a myoblast-specific super-enhancer (SE); as a seRNA it binds with a number of target genomic loci predominantly in trans. Further studies demonstrate that it interacts with Ddx5 to tether PAM SE to its inter-chromosomal targets Timp2 and Vim to activate the gene expression. Lastly, we show that PAM expression is increased in aging SCs, which leads to enhanced inter-chromosomal interaction and target genes upregulation. Altogether, our findings identify PAM as a previously unknown lncRNA that regulates both SC proliferation and aging through its trans gene regulatory activity.


Subject(s)
RNA, Long Noncoding , Satellite Cells, Skeletal Muscle , Cell Differentiation/genetics , Cell Proliferation/genetics , Muscle, Skeletal/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Satellite Cells, Skeletal Muscle/metabolism
4.
Stem Cell Reports ; 16(10): 2442-2458, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34534448

ABSTRACT

Skeletal muscle satellite cells (SCs) are stem cells responsible for muscle development and regeneration. Although CRISPR/Cas9 has been widely used, its application in endogenous SCs remains elusive. Here, we generate mice expressing Cas9 in SCs and achieve robust editing in juvenile SCs at the postnatal stage through AAV9-mediated short guide RNA (sgRNA) delivery. Additionally, we reveal that quiescent SCs are resistant to CRISPR/Cas9-mediated editing. As a proof of concept, we demonstrate efficient editing of master transcription factor (TF) Myod1 locus using the CRISPR/Cas9/AAV9-sgRNA system in juvenile SCs. Application on two key TFs, MYC and BCL6, unveils distinct functions in SC activation and muscle regeneration. Particularly, we reveal that MYC orchestrates SC activation through regulating 3D genome architecture. Its depletion results in strengthening of the topologically associating domain boundaries thus may affect gene expression. Altogether, our study establishes a platform for editing endogenous SCs that can be harnessed to elucidate the functionality of key regulators governing SC activities.


Subject(s)
Chromatin/metabolism , Genes, myc , Genome , MyoD Protein/metabolism , Proto-Oncogene Proteins c-bcl-6/metabolism , RNA, Guide, Kinetoplastida/metabolism , Satellite Cells, Skeletal Muscle/physiology , Animals , CRISPR-Cas Systems , Gene Editing/methods , Gene Expression Regulation , Mice , MyoD Protein/genetics , Nucleic Acid Conformation , Proto-Oncogene Proteins c-bcl-6/genetics , RNA, Guide, Kinetoplastida/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Front Physiol ; 11: 612230, 2020.
Article in English | MEDLINE | ID: mdl-33505317

ABSTRACT

Craniofacial morphogenesis depends on proper migration of neural crest cells and their interactions with placodes and other cell types. Hox genes provide positional information and are important in patterning the neural crest and pharyngeal arches (PAs) for coordinated formation of craniofacial structures. Hox genes are expressed in the surface ectoderm and epibranchial placodes, their roles in the pharyngeal epithelium and their downstream targets in regulating PA morphogenesis have not been established. We altered the Hox code in the pharyngeal region of the Hoxb3 Tg/+ mutant, in which Hoxb3 is driven to ectopically expressed in Hoxb2 domain in the second pharyngeal arch (PA2). In the transgenic mutant, ectopic Hoxb3 expression was restricted to the surface ectoderm, including the proximal epibranchial placodal region and the distal pharyngeal epithelium. The Hoxb3 Tg/+ mutants displayed hypoplasia of PA2, multiple neural crest-derived facial skeletal and nerve defects. Interestingly, we found that in the Hoxb3 Tg/+ mutant, expression of the Notch ligand Jag1 was specifically up-regulated in the ectodermal pharyngeal epithelial cells of PA2. By molecular experiments, we demonstrated that Hoxb3 could bind to an upstream genomic site S2 and directly regulate Jag1 expression. In the Hoxb3 Tg/+ mutant, elevated expression of Jag1 in the pharyngeal epithelium led to abnormal cellular interaction and deficiency of neural crest cells migrating into PA2. In summary, we showed that Hoxb3 regulates Jag1 expression and proposed a model of pharyngeal epithelium and neural crest interaction during pharyngeal arch development.

6.
Aging Cell ; 18(5): e12996, 2019 10.
Article in English | MEDLINE | ID: mdl-31325224

ABSTRACT

Epigenetic alterations occur in various cells and tissues during aging, but it is not known if such alterations are also associated with aging in skeletal muscle. Here, we examined the changes of a panel of histone modifications and found H3K27ac (an active enhancer mark) is markedly increased in aged human skeletal muscle tissues. Further analyses uncovered that the H3K27ac increase and enhancer activation are associated with the up-regulation of extracellular matrix (ECM) genes; this may result in alteration of the niche environment for skeletal muscle stem cells, also called satellite cells (SCs), which causes decreased myogenic potential and fibrogenic conversion of SCs. In mice, treatment of aging muscles with JQ1, an inhibitor of enhancer activation, inhibited the ECM up-regulation and fibrogenic conversion of SCs and restored their myogenic differentiation potential. Altogether, our findings not only uncovered a novel aspect of skeletal muscle aging that is associated with enhancer remodeling but also implicated JQ1 as a potential treatment approach for restoring SC function in aging muscle.


Subject(s)
Aging/metabolism , Epigenesis, Genetic , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Histones/chemistry , Histones/metabolism , Muscle, Skeletal/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Acetylation , Aging/drug effects , Animals , Azepines/pharmacology , Cell Differentiation/drug effects , Cell Line , Cellular Senescence/drug effects , Extracellular Matrix/drug effects , Extracellular Matrix/genetics , Fibroblasts/drug effects , Mice , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Satellite Cells, Skeletal Muscle/drug effects , Triazoles/pharmacology
7.
Methods Mol Biol ; 1668: 15-25, 2017.
Article in English | MEDLINE | ID: mdl-28842899

ABSTRACT

Transcriptional control of gene expression in skeletal muscle cell is involved in different processes ranging from muscle formation to regeneration. The identification of an increasing number of transcription factors, co-factors, and histone modifications has been greatly advanced by methods that allow studies of genome-wide chromatin-protein interactions. Chromatin immunoprecipitation with massively parallel DNA sequencing, or ChIP-seq, is a powerful tool for identifying binding sites of TFs/co-factors and histone modifications. The major steps of this technique involve immunoprecipitation of fragmented chromatin, followed by high-throughput sequencing to identify the protein bound regions genome-wide. Here, in this protocol, we will illustrate how the entire ChIP-seq is performed using global H3K27ac profiling in myoblast cells as an example.


Subject(s)
Chromatin Immunoprecipitation/methods , Chromatin/genetics , Muscle Fibers, Skeletal/metabolism , Transcription Factors/metabolism , Whole Genome Sequencing/methods , Animals , Binding Sites/genetics , Cell Line , Chromosome Mapping , Gene Library , Genome-Wide Association Study , Histones/genetics , Histones/metabolism , Mice , Transcription Factors/genetics
8.
Nucleic Acids Res ; 45(15): 8785-8805, 2017 Sep 06.
Article in English | MEDLINE | ID: mdl-28575289

ABSTRACT

Super-enhancers (SEs) are cis-regulatory elements enriching lineage specific key transcription factors (TFs) to form hotspots. A paucity of identification and functional dissection promoted us to investigate SEs during myoblast differentiation. ChIP-seq analysis of histone marks leads to the uncovering of SEs which remodel progressively during the course of differentiation. Further analyses of TF ChIP-seq enable the definition of SE hotspots co-bound by the master TF, MyoD and other TFs, among which we perform in-depth dissection for MyoD/FoxO3 interaction in driving the hotspots formation and SE activation. Furthermore, using Myogenin as a model locus, we elucidate the hierarchical and complex interactions among hotspots during the differentiation, demonstrating SE function is propelled by the physical and functional cooperation among hotspots. Finally, we show MyoD and FoxO3 are key in orchestrating the Myogenin hotspots interaction and activation. Altogether our results identify muscle-specific SEs and provide mechanistic insights into the functionality of SE.


Subject(s)
Cell Differentiation/genetics , Enhancer Elements, Genetic/physiology , Forkhead Box Protein O3/physiology , Muscle Development/genetics , MyoD Protein/physiology , Animals , Cells, Cultured , Forkhead Box Protein O3/metabolism , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Mice , MyoD Protein/metabolism , Myoblasts/physiology , Myogenin/genetics , Myogenin/metabolism , Protein Binding
9.
Cell Discov ; 3: 17002, 2017.
Article in English | MEDLINE | ID: mdl-28326190

ABSTRACT

Malat1 is one of the most abundant long non-coding RNAs in various cell types; its exact cellular function is still a matter of intense investigation. In this study we characterized the function of Malat1 in skeletal muscle cells and muscle regeneration. Utilizing both in vitro and in vivo assays, we demonstrate that Malat1 has a role in regulating gene expression during myogenic differentiation of myoblast cells. Specifically, we found that knockdown of Malat1 accelerates the myogenic differentiation in cultured cells. Consistently, Malat1 knockout mice display enhanced muscle regeneration after injury and deletion of Malat1 in dystrophic mdx mice also improves the muscle regeneration. Mechanistically, in the proliferating myoblasts, Malat1 recruits Suv39h1 to MyoD-binding loci, causing trimethylation of histone 3 lysine 9 (H3K9me3), which suppresses the target gene expression. Upon differentiation, the pro-myogenic miR-181a is increased and targets the nuclear Malat1 transcripts for degradation through Ago2-dependent nuclear RNA-induced silencing complex machinery; the Malat1 decrease subsequently leads to the destabilization of Suv39h1/HP1ß/HDAC1-repressive complex and displacement by a Set7-containing activating complex, which allows MyoD trans-activation to occur. Together, our findings identify a regulatory axis of miR-181a-Malat1-MyoD/Suv39h1 in myogenesis and uncover a previously unknown molecular mechanism of Malat1 action in gene regulation.

SELECTION OF CITATIONS
SEARCH DETAIL
...