Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
EMBO Rep ; 25(3): 1256-1281, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38429579

ABSTRACT

The plant homeodomain zinc-finger protein, PHF6, is a transcriptional regulator, and PHF6 germline mutations cause the X-linked intellectual disability (XLID) Börjeson-Forssman-Lehmann syndrome (BFLS). The mechanisms by which PHF6 regulates transcription and how its mutations cause BFLS remain poorly characterized. Here, we show genome-wide binding of PHF6 in the developing cortex in the vicinity of genes involved in central nervous system development and neurogenesis. Characterization of BFLS mice harbouring PHF6 patient mutations reveals an increase in embryonic neural stem cell (eNSC) self-renewal and a reduction of neural progenitors. We identify a panel of Ephrin receptors (EphRs) as direct transcriptional targets of PHF6. Mechanistically, we show that PHF6 regulation of EphR is impaired in BFLS mice and in conditional Phf6 knock-out mice. Knockdown of EphR-A phenocopies the PHF6 loss-of-function defects in altering eNSCs, and its forced expression rescues defects of BFLS mice-derived eNSCs. Our data indicate that PHF6 directly promotes Ephrin receptor expression to control eNSC behaviour in the developing brain, and that this pathway is impaired in BFLS.


Subject(s)
Epilepsy , Face/abnormalities , Fingers/abnormalities , Growth Disorders , Hypogonadism , Intellectual Disability , Mental Retardation, X-Linked , Obesity , Humans , Mice , Animals , Intellectual Disability/genetics , Repressor Proteins , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/metabolism , Epilepsy/genetics , Epilepsy/metabolism , Transcription Factors
2.
Curr Protoc ; 3(8): e877, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37638781

ABSTRACT

Adult stem cells play a critical role in the maintenance and repair of the organs in which they reside. However, their function is highly dependent on the crosstalk with their niche environment that changes during development and in disease states. The niche provides signals to stem cells to activate, proliferate, self-renew, or remain in quiescence. In skeletal muscle, the niche is perturbed in disease contexts such as aging, muscular dystrophies, and cachexia. Therefore, it is important to develop methods that permit the decoupling of niche-mediated from cell-intrinsic changes that occur in muscle stem cells (MuSCs) in development and disease contexts. With the purpose of determining the effect of the niche environment on the MuSC transcriptome, function, or health, we have coupled an allogeneic stem cell transplantation system, meaning the transplantation of MuSCs from a donor mouse into a recipient host mouse, with Switching Mechanism at 5' End of RNA Template (SMART-Seq) to quantify the effects of the niche on the MuSC transcriptome in vivo. Briefly, MuSCs are isolated from a GFP reporter donor mouse (Pax7-nGFP) and transplanted into the irradiated muscles of immunocompromised allogeneic hosts. The MuSCs are re-isolated by fluorescence-activated cell sorting (FACS) after three weeks of inhabiting the heterologous niche, defined as a niche that is different from their originating niche, and sequencing-ready libraries are created. This method allows for the direct comparison of the transcriptome of stem cells before and after transplantation into a host of a different age, disease status, or genetic background. This method can be used to accurately quantify the direct effect of the niche environment on the stem cell gene expression profile and to decouple cell-intrinsic versus niche-mediated alterations in the stem cell transcriptome. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol: Allogeneic muscle stem cell transplantation.


Subject(s)
Adult Stem Cells , Hematopoietic Stem Cell Transplantation , Animals , Mice , Humans , Muscle Fibers, Skeletal , Muscle, Skeletal , Tissue Donors
3.
Nat Commun ; 14(1): 535, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36726011

ABSTRACT

Adult stem cells are indispensable for tissue regeneration, but their function declines with age. The niche environment in which the stem cells reside plays a critical role in their function. However, quantification of the niche effect on stem cell function is lacking. Using muscle stem cells (MuSC) as a model, we show that aging leads to a significant transcriptomic shift in their subpopulations accompanied by locus-specific gain and loss of chromatin accessibility and DNA methylation. By combining in vivo MuSC transplantation and computational methods, we show that the expression of approximately half of all age-altered genes in MuSCs from aged male mice can be restored by exposure to a young niche environment. While there is a correlation between gene reversibility and epigenetic alterations, restoration of gene expression occurs primarily at the level of transcription. The stem cell niche environment therefore represents an important therapeutic target to enhance tissue regeneration in aging.


Subject(s)
Adult Stem Cells , Muscle, Skeletal , Male , Mice , Animals , Muscle, Skeletal/metabolism , Muscle Fibers, Skeletal , Stem Cells/metabolism , Aging/physiology
4.
FEBS J ; 290(5): 1267-1289, 2023 03.
Article in English | MEDLINE | ID: mdl-35029021

ABSTRACT

Muscle stem cells (MuSCs) are required for life-long muscle regeneration. In general, aging has been linked to a decline in the numbers and the regenerative potential of MuSCs. Muscle regeneration depends on the proper functioning of MuSCs, which is itself dependent on intricate interactions with its niche components. Aging is associated with both cell-intrinsic and niche-mediated changes, which can be the result of transcriptional, posttranscriptional, or posttranslational alterations in MuSCs or in the components of their niche. The interplay between cell intrinsic alterations in MuSCs and changes in the stem cell niche environment during aging and its impact on the number and the function of MuSCs is an important emerging area of research. In this review, we discuss whether the decline in the regenerative potential of MuSCs with age is the cause or the consequence of aging skeletal muscle. Understanding the effect of aging on MuSCs and the individual components of their niche is critical to develop effective therapeutic approaches to diminish or reverse the age-related defects in muscle regeneration.


Subject(s)
Muscle, Skeletal , Satellite Cells, Skeletal Muscle , Muscle, Skeletal/physiology , Stem Cells , Regeneration/physiology
5.
Bio Protoc ; 12(12)2022 Jun 20.
Article in English | MEDLINE | ID: mdl-35990589

ABSTRACT

Chromatin accessibility is a key determinant of gene expression that can be altered under different physiological and disease conditions. Skeletal muscle is made up of myofibers that are highly plastic and adaptive. Therefore, assessing the genome-wide chromatin state of myofibers under various conditions is very important to gain insight into the epigenetic state of myonuclei. The rigid nature of myofibers, as well as the low number of myonuclei that they contain, have rendered genome-wide studies with myofibers challenging. In recent years, ATAC-Seq from whole muscle and single nucleus ATAC-Seq have been performed. However, these techniques cannot distinguish between different fiber and cell types present in the muscle. In addition, due to the limited depth capacity obtained from single nucleus ATAC-Seq, an extensive comparative analysis cannot be performed. Here, we introduce a protocol where we combine the isolation of a single myofiber with OMNI ATAC-Seq. This protocol allows for genome-wide analysis of accessible chromatin regions of a selected single myofiber at a sufficient depth for comparative analysis under various physiological and disease conditions. This protocol can also allow for a specific myofiber to be selected, such as a regenerating myofiber. In the future, this protocol can help identify global changes in chromatin state under various conditions, as well as between different types of myofibers. Graphical abstract.

6.
Methods Mol Biol ; 2515: 343-354, 2022.
Article in English | MEDLINE | ID: mdl-35776362

ABSTRACT

The generation of new neurons in the adult brain throughout life is integral to brain plasticity and repair. Adult neural stem cells (aNSCs), present in the subventricular zone (SVZ) of the lateral ventricle wall and the subgranular zone (SGZ) of the hippocampal dentate gyrus, divide symmetrically or asymmetrically to maintain the stem cell pool or become committed progenitors and differentiate into various cell lineages. Depletion or dysregulation of aNSCs impairs proper brain connectivity and function and can contribute to several brain diseases including cognitive and neurodegenerative disorders and brain cancer. In this chapter, we present our optimized method to obtain and maintain reproducible neurosphere cultures from the adult mouse brain followed by evaluation of self-renewal using the extreme limiting dilution assay (ELDA) software. We use this assay routinely on aNSCs obtained from patient mouse models to generate log fraction plots and provide confidence intervals for all limiting dilution assay (LDA) data. At the same time, given the low number of NSCs required for the completion of the ELDA experiment, it is feasible to employ this approach to conduct high-content compound screening for therapeutic interventions aimed at enhancing the stem cell pool or combating a cohort of genetic and epigenetic disorders.


Subject(s)
Adult Stem Cells , Neural Stem Cells , Animals , Brain , Humans , Lateral Ventricles , Mice , Neurons
7.
Methods Cell Biol ; 170: 47-58, 2022.
Article in English | MEDLINE | ID: mdl-35811103

ABSTRACT

Brain tumor stem cells (BTSCs) are a rare population of self-renewing stem cells that are cultured as spheres and are often slow growing compared to other mammalian cell lines. Analysis of BTSC proteome requires careful handling as well as techniques that can be applied to small quantities of cell material. Subcellular fractionation is a widely used technique to assess protein localization. Although proteins are often destined to a defined cell compartment via a signal peptide such as mitochondrial or nuclear localization signals, the recruitment of a protein from one compartment to another can occur as a result of post-translational modification and/or structural variations in response to intracellular and extracellular stimuli. These events assign different functions to a protein making the study of protein localization a useful approach for better understanding of its role in disease progression. Sequential centrifugation remains a simple and versatile fractionation method for proteomic analysis. It can also be applied for diverse downstream applications such as multi-omics using pure nuclear fractions or metabolomic studies on isolated mitochondria. In this chapter, we describe our optimized protocol for subcellular fractionation of BTSC spheres in which we use a commercially available kit with additional centrifugation steps. We provide details on BTSC maintenance and handling, fractionation protocol and evaluation of fraction purity.


Subject(s)
Neoplastic Stem Cells , Proteomics , Animals , Brain/metabolism , Cell Fractionation/methods , Cell Nucleus/metabolism , Mammals/metabolism , Neoplastic Stem Cells/pathology , Proteome/metabolism , Proteomics/methods , Subcellular Fractions/metabolism
8.
STAR Protoc ; 3(3): 101554, 2022 09 16.
Article in English | MEDLINE | ID: mdl-35880130

ABSTRACT

Improper or aberrant protein-protein interactions can lead to severe human diseases including cancer. Here, we describe an adapted proximity ligation assay (PLA) protocol for the assessment of galectin-1-HOXA5 interaction in brain tumor stem cells (BTSCs). We detail the steps for culturing and preparation of BTSCs followed by PLA and detection of protein interactions in situ using fluorescent microscopy. This PLA protocol is optimized specifically for BTSCs and includes key controls for effective result analysis. For complete details on the use and execution of this protocol, please refer to Sharanek et al. (2021).


Subject(s)
Brain , Protein Interaction Mapping , Humans , Microscopy, Fluorescence/methods , Neoplastic Stem Cells , Protein Interaction Mapping/methods
9.
Elife ; 112022 02 21.
Article in English | MEDLINE | ID: mdl-35188098

ABSTRACT

Myofibers are the main components of skeletal muscle, which is the largest tissue in the body. Myofibers are highly adaptive and can be altered under different biological and disease conditions. Therefore, transcriptional and epigenetic studies on myofibers are crucial to discover how chromatin alterations occur in the skeletal muscle under different conditions. However, due to the heterogenous nature of skeletal muscle, studying myofibers in isolation proves to be a challenging task. Single-cell sequencing has permitted the study of the epigenome of isolated myonuclei. While this provides sequencing with high dimensionality, the sequencing depth is lacking, which makes comparisons between different biological conditions difficult. Here, we report the first implementation of single myofiber ATAC-Seq, which allows for the sequencing of an individual myofiber at a depth sufficient for peak calling and for comparative analysis of chromatin accessibility under various physiological and disease conditions. Application of this technique revealed significant differences in chromatin accessibility between resting and regenerating myofibers, as well as between myofibers from a mouse model of Duchenne Muscular Dystrophy (mdx) and wild-type (WT) counterparts. This technique can lead to a wide application in the identification of chromatin regulatory elements and epigenetic mechanisms in muscle fibers during development and in muscle-wasting diseases.


Subject(s)
Chromatin , Muscular Dystrophy, Duchenne , Animals , Chromatin/genetics , Chromatin Immunoprecipitation Sequencing , Mice , Mice, Inbred mdx , Muscle Fibers, Skeletal , Muscle, Skeletal
10.
Cell Rep ; 36(9): 109647, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34469737

ABSTRACT

Brain tumor stem cells (BTSCs) and intratumoral heterogeneity represent major challenges in glioblastoma therapy. Here, we report that the LGALS1 gene, encoding the carbohydrate binding protein, galectin1, is a key regulator of BTSCs and glioblastoma resistance to therapy. Genetic deletion of LGALS1 alters BTSC gene expression profiles and results in downregulation of gene sets associated with the mesenchymal subtype of glioblastoma. Using a combination of pharmacological and genetic approaches, we establish that inhibition of LGALS1 signaling in BTSCs impairs self-renewal, suppresses tumorigenesis, prolongs lifespan, and improves glioblastoma response to ionizing radiation in preclinical animal models. Mechanistically, we show that LGALS1 is a direct transcriptional target of STAT3 with its expression robustly regulated by the ligand OSM. Importantly, we establish that galectin1 forms a complex with the transcription factor HOXA5 to reprogram the BTSC transcriptional landscape. Our data unravel an oncogenic signaling pathway by which the galectin1/HOXA5 complex maintains BTSCs and promotes glioblastoma.


Subject(s)
Brain Neoplasms/metabolism , Galectin 1/metabolism , Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , Transcription, Genetic , Aged , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Calixarenes/pharmacology , Cell Line, Tumor , Cell Proliferation , Cell Self Renewal , ErbB Receptors/genetics , ErbB Receptors/metabolism , Galectin 1/antagonists & inhibitors , Galectin 1/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/radiotherapy , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Mice, SCID , Middle Aged , Mutation , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , Radiation Tolerance , Radiation-Sensitizing Agents/pharmacology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Xenograft Model Antitumor Assays
11.
Crit Rev Biochem Mol Biol ; 56(3): 284-300, 2021 06.
Article in English | MEDLINE | ID: mdl-33823731

ABSTRACT

Muscle stem cells (MuSCs) also called satellite cells are the building blocks of skeletal muscle, the largest tissue in the human body which is formed primarily of myofibers. While MuSCs are the principal cells that directly contribute to the formation of the muscle fibers, their ability to do so depends on critical interactions with a vast array of nonmyogenic cells within their niche environment. Therefore, understanding the nature of communication between MuSCs and their niche is of key importance to understand how the skeletal muscle is maintained and regenerated after injury. MuSCs are rare and therefore difficult to study in vivo within the context of their niche environment. The advent of single-cell technologies, such as switching mechanism at 5' end of the RNA template (SMART) and tagmentation based technologies using hyperactive transposase, afford the unprecedented opportunity to perform whole transcriptome and epigenome studies on rare cells within their niche environment. In this review, we will delve into how single-cell technologies can be applied to the study of MuSCs and muscle-resident niche cells and the impact this can have on our understanding of MuSC biology and skeletal muscle regeneration.


Subject(s)
Epigenome , Genome-Wide Association Study , Myoblasts, Skeletal/physiology , Regeneration , Single-Cell Analysis , Stem Cell Niche , Transcriptome , Animals , Humans
12.
EMBO Rep ; 21(12): e49499, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33047485

ABSTRACT

The function and maintenance of muscle stem cells (MuSCs) are tightly regulated by signals originating from their niche environment. Skeletal myofibers are a principle component of the MuSC niche and are in direct contact with the muscle stem cells. Here, we show that Myf6 establishes a ligand/receptor interaction between muscle stem cells and their associated muscle fibers. Our data show that Myf6 transcriptionally regulates a broad spectrum of myokines and muscle-secreted proteins in skeletal myofibers, including EGF. EGFR signaling blocks p38 MAP kinase-induced differentiation of muscle stem cells. Homozygous deletion of Myf6 causes a significant reduction in the ability of muscle to produce EGF, leading to a deregulation in EGFR signaling. Consequently, although Myf6-knockout mice are born with a normal muscle stem cell compartment, they undergo a progressive reduction in their stem cell pool during postnatal life due to spontaneous exit from quiescence. Taken together, our data uncover a novel role for Myf6 in promoting the expression of key myokines, such as EGF, in the muscle fiber which prevents muscle stem cell exhaustion by blocking their premature differentiation.


Subject(s)
Myogenic Regulatory Factors , Stem Cells , Animals , Cell Differentiation/genetics , Homozygote , Mice , Muscle, Skeletal , Myogenic Regulatory Factors/genetics , Sequence Deletion
13.
Nat Commun ; 11(1): 4116, 2020 08 17.
Article in English | MEDLINE | ID: mdl-32807793

ABSTRACT

Glioblastoma contains a rare population of self-renewing brain tumor stem cells (BTSCs) which are endowed with properties to proliferate, spur the growth of new tumors, and at the same time, evade ionizing radiation (IR) and chemotherapy. However, the drivers of BTSC resistance to therapy remain unknown. The cytokine receptor for oncostatin M (OSMR) regulates BTSC proliferation and glioblastoma tumorigenesis. Here, we report our discovery of a mitochondrial OSMR that confers resistance to IR via regulation of oxidative phosphorylation, independent of its role in cell proliferation. Mechanistically, OSMR is targeted to the mitochondrial matrix via the presequence translocase-associated motor complex components, mtHSP70 and TIM44. OSMR interacts with NADH ubiquinone oxidoreductase 1/2 (NDUFS1/2) of complex I and promotes mitochondrial respiration. Deletion of OSMR impairs spare respiratory capacity, increases reactive oxygen species, and sensitizes BTSCs to IR-induced cell death. Importantly, suppression of OSMR improves glioblastoma response to IR and prolongs lifespan.


Subject(s)
Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , Radiation, Ionizing , Receptors, Oncostatin M/metabolism , Animals , Cell Death/radiation effects , Cell Line, Tumor , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Cells, Cultured , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Male , Mice , Mice, SCID , NADH Dehydrogenase/genetics , NADH Dehydrogenase/metabolism , Neoplastic Stem Cells/radiation effects , Oncostatin M/metabolism , Oxidative Stress/radiation effects , Receptors, Oncostatin M/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/radiation effects
14.
Bio Protoc ; 10(4): e3525, 2020 Feb 20.
Article in English | MEDLINE | ID: mdl-33654749

ABSTRACT

Whole transcriptome analysis is a key method in biology that allows researchers to determine the effect a condition has on gene regulation. One difficulty in RNA sequencing of muscle is that traditional methods are performed on the whole muscle, but this captures non-myogenic cells that are part of the muscle. In order to analyze only the transcriptome of myofibers we combine single myofiber isolation with SMART-Seq to provide high resolution genome wide expression of a single myofiber.

15.
J Biol Chem ; 294(52): 20097-20108, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31753917

ABSTRACT

Skeletal muscle is a heterogeneous tissue. Individual myofibers that make up muscle tissue exhibit variation in their metabolic and contractile properties. Although biochemical and histological assays are available to study myofiber heterogeneity, efficient methods to analyze the whole transcriptome of individual myofibers are lacking. Here, we report on a single-myofiber RNA-sequencing (smfRNA-Seq) approach to analyze the whole transcriptome of individual myofibers by combining single-fiber isolation with Switching Mechanism at 5' end of RNA Template (SMART) technology. Using smfRNA-Seq, we first determined the genes that are expressed in the whole muscle, including in nonmyogenic cells. We also analyzed the differences in the transcriptome of myofibers from young and old mice to validate the effectiveness of this new method. Our results suggest that aging leads to significant changes in the expression of metabolic genes, such as Nos1, and structural genes, such as Myl1, in myofibers. We conclude that smfRNA-Seq is a powerful tool to study developmental, disease-related, and age-related changes in the gene expression profile of skeletal muscle.


Subject(s)
Gene Expression Profiling/methods , RNA, Messenger/metabolism , Aging , Animals , Cell Separation/methods , Gene Library , Genome , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , RNA, Messenger/chemistry , Sequence Analysis, RNA/methods , Single-Cell Analysis , Transcriptome
16.
Nucleic Acids Res ; 46(14): 7221-7235, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30016497

ABSTRACT

Muscle-specific transcription factor MyoD orchestrates the myogenic gene expression program by binding to short DNA motifs called E-boxes within myogenic cis-regulatory elements (CREs). Genome-wide analyses of MyoD cistrome by chromatin immnunoprecipitation sequencing shows that MyoD-bound CREs contain multiple E-boxes of various sequences. However, how E-box numbers, sequences and their spatial arrangement within CREs collectively regulate the binding affinity and transcriptional activity of MyoD remain largely unknown. Here, by an integrative analysis of MyoD cistrome combined with genome-wide analysis of key regulatory histones and gene expression data we show that the affinity landscape of MyoD is driven by multiple E-boxes, and that the overall binding affinity-and associated nucleosome positioning and epigenetic features of the CREs-crucially depend on the variant sequences and positioning of the E-boxes within the CREs. By comparative genomic analysis of single nucleotide polymorphism (SNPs) across publicly available data from 17 strains of laboratory mice, we show that variant sequences within the MyoD-bound motifs, but not their genome-wide counterparts, are under selection. At last, we show that the quantitative regulatory effect of MyoD binding on the nearby genes can, in part, be predicted by the motif composition of the CREs to which it binds. Taken together, our data suggest that motif numbers, sequences and their spatial arrangement within the myogenic CREs are important determinants of the cis-regulatory code of myogenic CREs.


Subject(s)
E-Box Elements/genetics , Muscle Development/genetics , MyoD Protein/genetics , MyoD Protein/metabolism , Transcription, Genetic/genetics , Transcriptional Activation/genetics , Animals , Base Sequence/genetics , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , Gene Expression/genetics , Gene Expression Regulation , Genome-Wide Association Study , Mice , Muscle Development/physiology , Nucleotide Motifs/genetics , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/genetics
17.
Nat Neurosci ; 19(6): 798-806, 2016 06.
Article in English | MEDLINE | ID: mdl-27110918

ABSTRACT

EGFRvIII-STAT3 signaling is important in glioblastoma pathogenesis. Here, we identified the cytokine receptor OSMR as a direct target gene of the transcription factor STAT3 in mouse astrocytes and human brain tumor stem cells (BTSCs). We found that OSMR functioned as an essential co-receptor for EGFRvIII. OSMR formed a physical complex with EGFRvIII, and depletion of OSMR impaired EGFRvIII-STAT3 signaling. Conversely, pharmacological inhibition of EGFRvIII phosphorylation inhibited the EGFRvIII-OSMR interaction and activation of STAT3. EGFRvIII-OSMR signaling in tumors operated constitutively, whereas EGFR-OSMR signaling in nontumor cells was synergistically activated by the ligands EGF and OSM. Finally, knockdown of OSMR strongly suppressed cell proliferation and tumor growth of mouse glioblastoma cells and human BTSC xenografts in mice, and prolonged the lifespan of these mice. Our findings identify OSMR as a critical regulator of glioblastoma tumor growth that orchestrates a feed-forward signaling mechanism with EGFRvIII and STAT3 to drive tumorigenesis.


Subject(s)
Brain Neoplasms/metabolism , Cell Transformation, Neoplastic/metabolism , Cytokines/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Signal Transduction/physiology , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Glioblastoma/pathology , Humans , Male , Mice, Transgenic , Neoplasm Transplantation/methods , STAT3 Transcription Factor/metabolism
18.
Skelet Muscle ; 5: 14, 2015.
Article in English | MEDLINE | ID: mdl-25949788

ABSTRACT

BACKGROUND: Adult skeletal muscle regeneration is a highly orchestrated process involving the activation and proliferation of satellite cells, an adult skeletal muscle stem cell. Activated satellite cells generate a transient amplifying progenitor pool of myoblasts that commit to differentiation and fuse into multinucleated myotubes. During regeneration, canonical Wnt signalling is activated and has been implicated in regulating myogenic lineage progression and terminal differentiation. METHODS: Here, we have undertaken a gene expression analysis of committed satellite cell-derived myoblasts to examine their ability to respond to canonical Wnt/ß-catenin signalling. RESULTS: We found that activation of canonical Wnt signalling induces follistatin expression in myoblasts and promotes myoblast fusion in a follistatin-dependent manner. In growth conditions, canonical Wnt/ß-catenin signalling prime myoblasts for myogenic differentiation by stimulating myogenin and follistatin expression. We further found that myogenin binds elements in the follistatin promoter and thus acts downstream of myogenin during differentiation. Finally, ectopic activation of canonical Wnt signalling in vivo promoted premature differentiation during muscle regeneration following acute injury. CONCLUSIONS: Together, these data reveal a novel mechanism by which myogenin mediates the canonical Wnt/ß-catenin-dependent activation of follistatin and induction of the myogenic differentiation process.

19.
Nat Protoc ; 8(8): 1525-34, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23845964

ABSTRACT

Chromatin immunoprecipitation coupled with ultra-high-throughput sequencing (ChIP-seq) is a widely used method for mapping the interactions of proteins with DNA. However, the requirements for ChIP-grade antibodies impede wider application of this method, and variations in results can be high owing to differences in affinity and cross-reactivity of antibodies. Therefore, we developed chromatin tandem affinity purification (ChTAP) as an effective alternative to ChIP. Through the use of affinity tags and reagents that are identical for all proteins investigated, ChTAP enables one to directly compare the binding between different transcription factors and to directly assess the background in control experiments. Thus, ChTAP-seq can be used to rapidly map the genome-wide binding of multiple DNA-binding proteins in a wide range of cell types. ChTAP can be completed in 3-4 d, starting from cross-linking of chromatin to purification of ChIP DNA.


Subject(s)
Chromatin/metabolism , Chromatography, Affinity/methods , DNA-Binding Proteins/metabolism , Binding Sites , Chromatin Immunoprecipitation , HEK293 Cells , Humans , Sonication
20.
Cell Metab ; 17(2): 210-24, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23395168

ABSTRACT

Brown adipose tissue (BAT) is an energy-dispensing thermogenic tissue that plays an important role in balancing energy metabolism. Lineage-tracing experiments indicate that brown adipocytes are derived from myogenic progenitors during embryonic development. However, adult skeletal muscle stem cells (satellite cells) have long been considered uniformly determined toward the myogenic lineage. Here, we report that adult satellite cells give rise to brown adipocytes and that microRNA-133 regulates the choice between myogenic and brown adipose determination by targeting the 3'UTR of Prdm16. Antagonism of microRNA-133 during muscle regeneration increases uncoupled respiration, glucose uptake, and thermogenesis in local treated muscle and augments whole-body energy expenditure, improves glucose tolerance, and impedes the development of diet-induced obesity. Finally, we demonstrate that miR-133 levels are downregulated in mice exposed to cold, resulting in de novo generation of satellite cell-derived brown adipocytes. Therefore, microRNA-133 represents an important therapeutic target for the treatment of obesity.


Subject(s)
Adipose Tissue, Brown/cytology , DNA-Binding Proteins/metabolism , MicroRNAs/metabolism , Satellite Cells, Skeletal Muscle/cytology , Transcription Factors/metabolism , 3' Untranslated Regions/genetics , Adipocytes, Brown/cytology , Adipose Tissue, Brown/metabolism , Animals , Base Sequence , Cell Differentiation/genetics , Cell Lineage/genetics , Cold Temperature , Down-Regulation/genetics , Energy Metabolism , Glucose Tolerance Test , Mice , MicroRNAs/genetics , Molecular Sequence Data , Multipotent Stem Cells/cytology , Regeneration/genetics , Satellite Cells, Skeletal Muscle/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...