85 results on '"Vittorio Sartorelli"'
Search Results
2. Protocols to generate and isolate mouse myogenic progenitors both in vitro and in vivo
- Author
-
Mamduh Khateb, Xuesong Feng, Stefania Dell’Orso, and Vittorio Sartorelli
- Subjects
General Immunology and Microbiology ,General Neuroscience ,General Biochemistry, Genetics and Molecular Biology - Abstract
Mouse embryonic stem cells (mESCs) can be directed to acquire cell-lineage-specific genetic programs and phenotypes by stepwise exposure to defined factors, allowing the development of in vitro models for studying disease and tissue generation. In this protocol, we describe the use of cultured mESCs to generate presomitic-like mesoderm cells undergoing further specification towards myogenic progenitors (MPs). Further, we describe here a procedure to obtain, dissect, and fluorescence-activated cell sorting (FACS)-isolate somitic cells from genetically labeled Pax7
- Published
- 2022
3. Decision letter: Human DUX4 and mouse Dux interact with STAT1 and broadly inhibit interferon-stimulated gene induction
- Author
-
Vittorio Sartorelli
- Published
- 2022
- Full Text
- View/download PDF
4. Integrating single-cell transcriptomes, chromatin accessibility, and multiomics analysis of mesoderm-induced embryonic stem cells
- Author
-
Kyung Dae Ko, Kan Jiang, Stefania Dell’Orso, and Vittorio Sartorelli
- Subjects
General Immunology and Microbiology ,General Neuroscience ,General Biochemistry, Genetics and Molecular Biology - Published
- 2023
- Full Text
- View/download PDF
5. The JAK-STAT pathway at 30: Much learned, much more to do
- Author
-
Rachael L. Philips, Yuxin Wang, HyeonJoo Cheon, Yuka Kanno, Massimo Gadina, Vittorio Sartorelli, Curt M. Horvath, James E. Darnell, George R. Stark, and John J. O’Shea
- Subjects
Mammals ,STAT Transcription Factors ,Animals ,COVID-19 ,Cytokines ,Humans ,Interferons ,General Biochemistry, Genetics and Molecular Biology ,Article ,Janus Kinases ,Signal Transduction - Abstract
The discovery of the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway arose from investigations of how cells respond to interferons (IFNs), revealing a paradigm in cell signaling conserved from slime molds to mammals. These discoveries revealed mechanisms underlying rapid gene expression mediated by a wide variety of extracellular polypeptides including cytokines, interleukins, and related factors. This knowledge has provided numerous insights into human disease, from immune deficiencies to cancer, and was rapidly translated to new drugs for autoimmune, allergic, and infectious diseases, including COVID-19. Despite these advances, major challenges and opportunities remain.
- Published
- 2022
6. FACS-Isolation and Culture of Fibro-Adipogenic Progenitors and Muscle Stem Cells from Unperturbed and Injured Mouse Skeletal Muscle
- Author
-
Vittorio Sartorelli, James M. Simone, and Giulia Riparini
- Subjects
Mice ,Adipogenesis ,General Immunology and Microbiology ,Stem Cells ,General Chemical Engineering ,General Neuroscience ,Animals ,Cell Differentiation ,Flow Cytometry ,Muscle, Skeletal ,Fibrosis ,General Biochemistry, Genetics and Molecular Biology - Abstract
Fibro-adipogenic progenitor cells (FAPs) are a population of skeletal muscle-resident mesenchymal stromal cells (MSCs) capable of differentiating along fibrogenic, adipogenic, osteogenic, or chondrogenic lineage. Together with muscle stem cells (MuSCs), FAPs play a critical role in muscle homeostasis, repair, and regeneration, while actively maintaining and remodeling the extracellular matrix (ECM). In pathological conditions, such as chronic damage and muscular dystrophies, FAPs undergo aberrant activation and differentiate into collagen-producing fibroblasts and adipocytes, leading to fibrosis and intramuscular fatty infiltration. Thus, FAPs play a dual role in muscle regeneration, either by sustaining MuSC turnover and promoting tissue repair or contributing to fibrotic scar formation and ectopic fat infiltrates, which compromise the integrity and function of the skeletal muscle tissue. A proper purification of FAPs and MuSCs is a prerequisite for understanding the biological role of these cells in physiological as well as in pathological conditions. Here, we describe a standardized method for the simultaneous isolation of FAPs and MuSCs from limb muscles of adult mice using fluorescence-activated cell sorting (FACS). The protocol describes in detail the mechanical and enzymatic dissociation of mononucleated cells from whole limb muscles and injured tibialis anterior (TA) muscles. FAPs and MuSCs are subsequently isolated using a semi-automated cell sorter to obtain pure cell populations. We additionally describe an optimized method for culturing quiescent and activated FAPs and MuSCs, either alone or in coculture conditions.
- Published
- 2022
- Full Text
- View/download PDF
7. FoxO maintains a genuine muscle stem-cell quiescent state until geriatric age
- Author
-
Stephen R. Brooks, Hong-Wei Sun, Vittorio Sartorelli, Kan Jiang, Marco Sandri, Laura Ortet, Stefania Dell'Orso, Srikanth Ravichandran, Sonia Alonso-Martin, Aster H. Juan, Laura García-Prat, Victoria Moiseeva, Marta Flández, Eusebio Perdiguero, Vanessa Ruiz-Bonilla, Pura Muñoz-Cánoves, Elena Rebollo, Mercè Jardí, Antonio Musarò, Xiaotong Hong, Antonio del Sol, Silvia Campanario, Ministerio de Economía y Competitividad (España), Ministerio de Ciencia, Innovación y Universidades (España), Agencia Estatal de Investigación (España), European Commission, Foundation for the National Institutes of Health, and Fundación Severo Ochoa
- Subjects
aging ,satellite cells ,foxO ,stem-cell ,Male ,Antigens, CD34 ,Cell Cycle Proteins ,Mice, SCID ,0302 clinical medicine ,Cell Self Renewal ,Stem Cell Niche ,Cells, Cultured ,Cellular Senescence ,Mice, Knockout ,0303 health sciences ,Forkhead Box Protein O1 ,Forkhead Box Protein O3 ,Age Factors ,Quiescent state ,Forkhead Transcription Factors ,Cell biology ,Phenotype ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Signal Transduction ,Myogenic differentiation ,Satellite Cells, Skeletal Muscle ,education ,Biology ,Cardiotoxins ,03 medical and health sciences ,Muscle stem cells ,medicine ,Animals ,Regeneration ,Muscle, Skeletal ,Protein kinase B ,Cell Proliferation ,030304 developmental biology ,Skeletal muscle ,Cell Biology ,Mice, Inbred C57BL ,Young age ,Gene Expression Regulation ,Ageing ,Proto-Oncogene Proteins c-akt ,Homeostasis ,Muscle stem cell - Abstract
Tissue regeneration declines with ageing but little is known about whether this arises from changes in stem-cell heterogeneity. Here, in homeostatic skeletal muscle, we identify two quiescent stem-cell states distinguished by relative CD34 expression: CD34High, with stemness properties (genuine state), and CD34Low, committed to myogenic differentiation (primed state). The genuine-quiescent state is unexpectedly preserved into later life, succumbing only in extreme old age due to the acquisition of primed-state traits. Niche-derived IGF1-dependent Akt activation debilitates the genuine stem-cell state by imposing primed-state features via FoxO inhibition. Interventions to neutralize Akt and promote FoxO activity drive a primed-to-genuine state conversion, whereas FoxO inactivation deteriorates the genuine state at a young age, causing regenerative failure of muscle, as occurs in geriatric mice. These findings reveal transcriptional determinants of stem-cell heterogeneity that resist ageing more than previously anticipated and are only lost in extreme old age, with implications for the repair of geriatric muscle., The authors acknowledge funding from MINECO-Spain (grant no. RTI2018-096068), ERC2016-AdG-741966, LaCaixa-HEALTH-HR17-00040, MDA, UPGRADE-H2020-825825, AFM and DPP-Spain to P.M.-C; María-de-Maeztu-Program for Units of Excellence to UPF (grant no. MDM-2014-0370) and the Severo-Ochoa-Program for Centers of Excellence to CNIC (grant no. SEV-2015-0505). This work was also supported by NIAMS IRP through NIH grants nos AR041126 and AR041164 to V.S. and utilized computational resources of the NIH HPC Biowulf cluster (http://hpc.nih.gov); ASI, Ricerca Finalizzata, Ateneo Sapienza to A.M.; AIRC (grant no. 23257); ASI (grant no. MARS-PRE, DC-VUM-2017-006); H2020-MSCA-RISE-2014 (645648) to M.S. and a FNR core grant (grant no. C15/BM/10397420) to A.d.S. L.G.P. was partially supported by an FPI fellowship and an EMBO fellowship (grant no. ALTF 420-2017); and S.C., X.H. and V.M. by FI, Severo-Ochoa and PFI Fellowships (Spain), respectively.
- Published
- 2020
- Full Text
- View/download PDF
8. Enhancer RNAs are an important regulatory layer of the epigenome
- Author
-
Vittorio Sartorelli and Shannon M. Lauberth
- Subjects
RNA, Untranslated ,Transcription, Genetic ,Enhancer Elements ,1.1 Normal biological development and functioning ,Biophysics ,Computational biology ,Biology ,Medical and Health Sciences ,Article ,Epigenome ,03 medical and health sciences ,0302 clinical medicine ,Genetic ,Underpinning research ,Structural Biology ,Transcription (biology) ,Neoplasms ,Genetics ,Animals ,Humans ,Enhancer ,Molecular Biology ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,Rna processing ,Extramural ,Human Genome ,Untranslated ,RNA ,Biological Sciences ,Chromatin ,Enhancer Elements, Genetic ,Gene Expression Regulation ,Chemical Sciences ,Generic health relevance ,Transcription ,030217 neurology & neurosurgery ,Biogenesis ,Developmental Biology - Abstract
Noncoding RNAs (ncRNAs) direct a remarkable number of diverse functions in development and disease through their regulation of transcription, RNA processing and translation. Leading the charge in the RNA revolution is a class of ncRNAs that are synthesized at active enhancers, called enhancer RNAs (eRNAs). Here, we review recent insights into the biogenesis of eRNAs and the mechanisms underlying their multifaceted functions and consider how these findings could inform future investigations into enhancer transcription and eRNA function.
- Published
- 2020
- Full Text
- View/download PDF
9. Transcriptomics, regulatory syntax, and enhancer identification in mesoderm-induced ESCs at single-cell resolution
- Author
-
Mamduh Khateb, Jelena Perovanovic, Kyung Dae Ko, Kan Jiang, Xuesong Feng, Natalia Acevedo-Luna, Jérome Chal, Veronica Ciuffoli, Pavol Genzor, James Simone, Astrid D. Haase, Olivier Pourquié, Stefania Dell’Orso, and Vittorio Sartorelli
- Subjects
Mesoderm ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Regulatory Sequences, Nucleic Acid ,Transcriptome ,General Biochemistry, Genetics and Molecular Biology ,Embryonic Stem Cells - Abstract
Embryonic stem cells (ESCs) can adopt lineage-specific gene-expression programs by stepwise exposure to defined factors, resulting in the generation of functional cell types. Bulk and single-cell-based assays were employed to catalog gene expression, histone modifications, chromatin conformation, and accessibility transitions in ESC populations and individual cells acquiring a presomitic mesoderm fate and undergoing further specification toward myogenic and neurogenic lineages. These assays identified cis-regulatory regions and transcription factors presiding over gene-expression programs occurring at defined ESC transitions and revealed the presence of heterogeneous cell populations within discrete ESC developmental stages. The datasets were employed to identify previously unappreciated genomic elements directing the initial activation of Pax7 and myogenic and neurogenic gene-expression programs. This study provides a resource for the discovery of genomic and transcriptional features of pluripotent, mesoderm-induced ESCs and ESC-derived cell lineages.
- Published
- 2022
10. Transcriptomics, Regulatory Syntax, and Enhancer Identification in Heterogenous Populations of Mesoderm-Induced ESCs at Single-Cell Resolution
- Author
-
Mamduh Khateb, Jelena Perovanovic, Kyung Dae Ko, Kan Jiang, Xuesong Feng, Natalia Acevedo-Luna, Jérome Chal, Veronica Ciuffoli, Pavol Genzor, James Simone, Astrid D. Haase, Olivier Pourquié, Stefania Dell’Orso, and Vittorio Sartorelli
- Subjects
embryonic structures - Abstract
SUMMARYESCs can adopt lineage-specific gene expression programs by stepwise exposure to defined factors, resulting in the generation of functional cell types. Bulk and single cell-based assays were employed to catalogue gene expression, histone modifications, chromatin conformation, and accessibility transitions in ESC populations and individual cells acquiring a presomitic mesoderm fate and undergoing further lineage specification. These assays identified cis-regulatory regions and transcription factors presiding gene expression programs occurring at defined ESC transitions and revealed the presence of heterogeneous cell populations within discrete ESC developmental stages. The datasets were employed to identify previously unappreciated genomic elements directing the initial activation of Pax7 and myogenic and neurogenic gene expression programs. This study provides a resource for the discovery of genomic and transcriptional features of pluripotent, mesoderm-induced ESCs, and ESCs-derived cell lineages.
- Published
- 2022
- Full Text
- View/download PDF
11. Redundant mechanisms driven independently by RUNX1 and GATA2 for hematopoietic development
- Author
-
Vittorio Sartorelli, Elizabeth Broadbridge, Ursula Harper, Stephen Wincovitch, Paul P. Liu, Erica Bresciani, Stefania Dell'Orso, Martha Kirby, Tao Zhen, Raman Sood, Erika Mijin Kwon Kim, Blake Carrington, Victoria Sanchez Guzman, Kevin Bishop, and Kai Yu
- Subjects
Hemangioblasts ,Population ,Biology ,chemistry.chemical_compound ,Mice ,hemic and lymphatic diseases ,Animals ,education ,Zebrafish ,Hemogenic endothelium ,education.field_of_study ,Hematopoietic Tissue ,GATA2 ,Hematology ,Zebrafish Proteins ,biology.organism_classification ,Hematopoietic Stem Cells ,Cell biology ,Hematopoiesis ,GATA2 Transcription Factor ,Haematopoiesis ,RUNX1 ,chemistry ,embryonic structures ,Core Binding Factor Alpha 2 Subunit ,Stem cell - Abstract
RUNX1 is essential for the generation of hematopoietic stem cells (HSCs).Runx1null mouse embryos lack definitive hematopoiesis and die in mid-gestation. However, even though zebrafish embryos with arunx1W84X mutation have defects in early definitive hematopoiesis, somerunx1W84X/W84Xembryos can develop to fertile adults with blood cells of multi-lineages, raising the possibility that HSCs can emerge without RUNX1. Here, using three new zebrafishrunx1-/-lines we uncovered the compensatory mechanism forrunx1-independent hematopoiesis. We show that, in the absence of a functionalrunx1, acd41-GFP+population of hematopoietic precursors still emerge from the hemogenic endothelium and can colonize the hematopoietic tissues of the mutant embryos. Single-cell RNA sequencing of thecd41-GFP+cells identified a set ofrunx1-/--specific signature genes during hematopoiesis. Significantly,gata2b, which normally acts upstream ofrunx1for the generation of HSCs, was increased in thecd41-GFP+cells inrunx1- /-embryos. Interestingly, genetic inactivation of bothgata2band its paralog,gata2a, did not affect hematopoiesis. However, knocking outrunx1and any three of the four alleles ofgata2aandgata2babolished definitive hematopoiesis.Gata2expression was also upregulated in hematopoietic cells inRunx1-/-mice, suggesting the compensatory mechanism is conserved. Our findings indicate that RUNX1 and GATA2 serve redundant roles for HSC production, acting as each other’s safeguard.Key pointsExistence of RUNX1-independent mechanisms for the generation of HSCs and the development of functional definitive hematopoietic cellsGATA2 and RUNX1 functionally complement each other for their respective roles during hematopoiesis
- Published
- 2020
12. Correction: Single cell analysis of adult mouse skeletal muscle stem cells in homeostatic and regenerative conditions (doi: 10.1242/dev.174177)
- Author
-
Aster H. Juan, Vittorio Sartorelli, Gustavo Gutierrez-Cruz, Kyung-Dae Ko, Xuesong Feng, Jelena Perovanovic, Faiza Naz, and Stefania Dell'Orso
- Subjects
0303 health sciences ,Skeletal muscle ,Biology ,Cell biology ,03 medical and health sciences ,Techniques and Resources ,0302 clinical medicine ,medicine.anatomical_structure ,Single-cell analysis ,medicine ,Stem cell ,Molecular Biology ,030217 neurology & neurosurgery ,Homeostasis ,030304 developmental biology ,Developmental Biology - Abstract
Dedicated stem cells ensure postnatal growth, repair and homeostasis of skeletal muscle. Following injury, muscle stem cells (MuSCs) exit from quiescence and divide to reconstitute the stem cell pool and give rise to muscle progenitors. The transcriptomes of pooled MuSCs have provided a rich source of information for describing the genetic programs of distinct static cell states; however, bulk microarray and RNA sequencing provide only averaged gene expression profiles, blurring the heterogeneity and developmental dynamics of asynchronous MuSC populations. Instead, the granularity required to identify distinct cell types, states, and their dynamics can be afforded by single cell analysis. We were able to compare the transcriptomes of thousands of MuSCs and primary myoblasts isolated from homeostatic or regenerating muscles by single cell RNA sequencing. Using computational approaches, we could reconstruct dynamic trajectories and place, in a pseudotemporal manner, the transcriptomes of individual MuSC within these trajectories. This approach allowed for the identification of distinct clusters of MuSCs and primary myoblasts with partially overlapping but distinct transcriptional signatures, as well as the description of metabolic pathways associated with defined MuSC states.
- Published
- 2019
- Full Text
- View/download PDF
13. Single cell analysis of adult mouse skeletal muscle stem cells in homeostatic and regenerative conditions
- Author
-
Vittorio Sartorelli, Kyung-Dae Ko, Faiza Naz, Gustavo Gutierrez-Cruz, Aster H. Juan, Stefania Dell'Orso, Xuesong Feng, and Jelena Perovanovic
- Subjects
Cell type ,Cell ,Cell Separation ,Biology ,Muscle Development ,Transcriptome ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Single-cell analysis ,medicine ,Myocyte ,Animals ,Cluster Analysis ,Homeostasis ,Regeneration ,RNA-Seq ,Progenitor cell ,Muscle, Skeletal ,Molecular Biology ,030304 developmental biology ,Oligonucleotide Array Sequence Analysis ,0303 health sciences ,Sequence Analysis, RNA ,Stem Cells ,Skeletal muscle ,Computational Biology ,Correction ,Genomics ,Flow Cytometry ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Leukocytes, Mononuclear ,Stem cell ,Single-Cell Analysis ,030217 neurology & neurosurgery ,Software ,Developmental Biology - Abstract
Dedicated stem cells ensure post-natal growth, repair, and homeostasis of skeletal muscle. Following injury, muscle stem cells (MuSCs) exit from quiescence and divide to reconstitute the stem cell pool and give rise to muscle progenitors. The transcriptomes of pooled MuSCs have provided a rich source of information for describing the genetic programs of distinct static cell states; however, bulk microarray and RNA-seq provide only averaged gene expression profiles, blurring the heterogeneity and developmental dynamics of asynchronous MuSC populations. Instead, the granularity required to identify distinct cell types, states, and their dynamics can be afforded by single-cell analysis. We were able to compare the transcriptomes of thousands of MuSCs and primary myoblasts isolated from homeostatic or regenerating muscles by single-cell RNA- sequencing. Using computational approaches, we could reconstruct dynamic trajectories and place, in a pseudotemporal manner, the transcriptomes of individual MuSC within these trajectories. This approach allowed for the identification of distinct clusters of MuSCs and primary myoblasts with partially overlapping but distinct transcriptional signatures, as well as the description of metabolic pathways associated with defined MuSC states.
- Published
- 2018
14. Metabolic Reprogramming of Stem Cell Epigenetics
- Author
-
Vittorio Sartorelli, Stephen Dalton, Tim S Cliff, and James G. Ryall
- Subjects
Pluripotent Stem Cells ,Transcription, Genetic ,Somatic cell ,Cellular differentiation ,Citric Acid Cycle ,Induced Pluripotent Stem Cells ,Biology ,Article ,Oxidative Phosphorylation ,Epigenesis, Genetic ,Histones ,Histone demethylation ,Genetics ,Animals ,Humans ,Cell Lineage ,Epigenetics ,Induced pluripotent stem cell ,Embryonic Stem Cells ,Stem Cells ,Proteins ,Cell Differentiation ,Cell Biology ,Cellular Reprogramming ,Embryonic stem cell ,Carbon ,3. Good health ,Cell biology ,Oxygen ,Gene Expression Regulation ,Molecular Medicine ,Stem cell ,Glycolysis ,Reprogramming - Abstract
For many years, stem cell metabolism was viewed as a byproduct of cell fate status rather than an active regulatory mechanism; however, there is now a growing appreciation that metabolic pathways influence epigenetic changes associated with lineage commitment, specification, and self-renewal. Here we review how metabolites generated during glycolytic and oxidative processes are utilized in enzymatic reactions leading to epigenetic modifications and transcriptional regulation. We discuss how "metabolic reprogramming" contributes to global epigenetic changes in the context of naive and primed pluripotent states, somatic reprogramming, and hematopoietic and skeletal muscle tissue stem cells, and we discuss the implications for regenerative medicine.
- Published
- 2015
- Full Text
- View/download PDF
15. ATP Citrate Lyase: A New Player Linking Skeletal Muscle Metabolism and Epigenetics
- Author
-
Vittorio Sartorelli and Haisen Li
- Subjects
0301 basic medicine ,ATP citrate lyase ,Endocrinology, Diabetes and Metabolism ,Cellular differentiation ,MyoD ,Histones ,03 medical and health sciences ,Histone H3 ,Endocrinology ,medicine ,Regeneration ,Epigenetics ,Muscle, Skeletal ,biology ,Chemistry ,Skeletal muscle ,Acetylation ,Cell Differentiation ,musculoskeletal system ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Histone ,biology.protein ,ATP Citrate (pro-S)-Lyase - Abstract
Intermediates generated in several metabolic processes are used to regulate transcription through covalent histone and DNA modifications. In Cell Reports, Das et al. show that acetyl-coenzyme A (acetyl-CoA) generated by ATP citrate lyase (ACL) is utilized to acetylate histone H3 at MyoD regulatory regions, resulting in increased MyoD expression and improved muscle regeneration after injury.
- Published
- 2018
16. Super-enhancers delineate disease-associated regulatory nodes in T cells
- Author
-
Kan Jiang, Stephen C. J. Parker, Michael R. Erdos, Massimo Gadina, Francis S. Collins, Yasuko Furumoto, John J. O'Shea, Vittorio Sartorelli, Yuka Kanno, Golnaz Vahedi, Zhonghui Tang, Nicholas P. Restifo, Yijun Ruan, Sean Davis, and Rahul Roychoudhuri
- Subjects
RNA, Untranslated ,Transcription, Genetic ,Cellular differentiation ,medicine.medical_treatment ,P300-CBP Transcription Factors ,Biology ,Article ,Arthritis, Rheumatoid ,Mice ,Piperidines ,medicine ,Animals ,Cell Lineage ,Genetic Predisposition to Disease ,Pyrroles ,p300-CBP Transcription Factors ,IL-2 receptor ,Regulation of gene expression ,Genetics ,Multidisciplinary ,Janus kinase 3 ,Janus Kinase 3 ,Cell Differentiation ,T-Lymphocytes, Helper-Inducer ,Mice, Inbred C57BL ,Basic-Leucine Zipper Transcription Factors ,Enhancer Elements, Genetic ,Pyrimidines ,Cytokine ,Gene Expression Regulation ,Spatiotemporal gene expression ,Janus kinase - Abstract
Enhancers regulate spatiotemporal gene expression and impart cell-specific transcriptional outputs that drive cell identity. Super-enhancers (SEs), also known as stretch-enhancers, are a subset of enhancers especially important for genes associated with cell identity and genetic risk of disease. CD4(+) T cells are critical for host defence and autoimmunity. Here we analysed maps of mouse T-cell SEs as a non-biased means of identifying key regulatory nodes involved in cell specification. We found that cytokines and cytokine receptors were the dominant class of genes exhibiting SE architecture in T cells. Nonetheless, the locus encoding Bach2, a key negative regulator of effector differentiation, emerged as the most prominent T-cell SE, revealing a network in which SE-associated genes critical for T-cell biology are repressed by BACH2. Disease-associated single-nucleotide polymorphisms for immune-mediated disorders, including rheumatoid arthritis, were highly enriched for T-cell SEs versus typical enhancers or SEs in other cell lineages. Intriguingly, treatment of T cells with the Janus kinase (JAK) inhibitor tofacitinib disproportionately altered the expression of rheumatoid arthritis risk genes with SE structures. Together, these results indicate that genes with SE architecture in T cells encompass a variety of cytokines and cytokine receptors but are controlled by a 'guardian' transcription factor, itself endowed with an SE. Thus, enumeration of SEs allows the unbiased determination of key regulatory nodes in T cells, which are preferentially modulated by pharmacological intervention.
- Published
- 2015
- Full Text
- View/download PDF
17. The NAD+-Dependent SIRT1 Deacetylase Translates a Metabolic Switch into Regulatory Epigenetics in Skeletal Muscle Stem Cells
- Author
-
Assia Derfoul, Stefania Dell'Orso, Vittorio Sartorelli, Marcella Fulco, Gustavo Gutierrez-Cruz, Daphney Clermont, Miroslav Koulnis, James G. Ryall, Xuesong Feng, Aster H. Juan, and Hossein Zare
- Subjects
Epigenesis, Genetic ,Histones ,Mice ,Sirtuin 1 ,Genetics ,medicine ,Animals ,Myocyte ,Epigenetics ,Muscle, Skeletal ,biology ,Myogenesis ,Stem Cells ,Skeletal muscle ,Acetylation ,Cell Biology ,NAD ,Cell biology ,medicine.anatomical_structure ,Biochemistry ,biology.protein ,Molecular Medicine ,Histone deacetylase ,Stem cell ,Reprogramming - Abstract
SummaryStem cells undergo a shift in metabolic substrate utilization during specification and/or differentiation, a process that has been termed metabolic reprogramming. Here, we report that during the transition from quiescence to proliferation, skeletal muscle stem cells experience a metabolic switch from fatty acid oxidation to glycolysis. This reprogramming of cellular metabolism decreases intracellular NAD+ levels and the activity of the histone deacetylase SIRT1, leading to elevated H4K16 acetylation and activation of muscle gene transcription. Selective genetic ablation of the SIRT1 deacetylase domain in skeletal muscle results in increased H4K16 acetylation and deregulated activation of the myogenic program in SCs. Moreover, mice with muscle-specific inactivation of the SIRT1 deacetylase domain display reduced myofiber size, impaired muscle regeneration, and derepression of muscle developmental genes. Overall, these findings reveal how metabolic cues can be mechanistically translated into epigenetic modifications that regulate skeletal muscle stem cell biology.
- Published
- 2015
- Full Text
- View/download PDF
18. The Elongation Factor Spt6 Maintains ESC Pluripotency by Controlling Super-Enhancers and Counteracting Polycomb Proteins
- Author
-
Aster H. Juan, Vittorio Sartorelli, Pei-Fang Tsai, Hossein Zare, Stefania Dell'Orso, A. Hongjun Wang, and Kyung Dae Ko
- Subjects
0301 basic medicine ,Down-Regulation ,macromolecular substances ,Biology ,Cell Line ,Histones ,03 medical and health sciences ,Mice ,Nucleosome ,Animals ,Enhancer of Zeste Homolog 2 Protein ,Enhancer ,Molecular Biology ,EZH2 ,Polycomb Repressive Complex 2 ,Promoter ,Acetylation ,Mouse Embryonic Stem Cells ,Cell Biology ,Molecular biology ,Chromatin ,Elongation factor ,030104 developmental biology ,Histone ,Enhancer Elements, Genetic ,biology.protein ,PRC2 ,Transcription Factors - Abstract
Summary Spt6 coordinates nucleosome dis- and re-assembly, transcriptional elongation, and mRNA processing. Here, we report that depleting Spt6 in embryonic stem cells (ESCs) reduced expression of pluripotency factors, increased expression of cell-lineage-affiliated developmental regulators, and induced cell morphological and biochemical changes indicative of ESC differentiation. Selective downregulation of pluripotency factors upon Spt6 depletion may be mechanistically explained by its enrichment at ESC super-enhancers, where Spt6 controls histone H3K27 acetylation and methylation and super-enhancer RNA transcription. In ESCs, Spt6 interacted with the PRC2 core subunit Suz12 and prevented H3K27me3 accumulation at ESC super-enhancers and associated promoters. Biochemical as well as functional experiments revealed that Spt6 could compete for binding of the PRC2 methyltransferase Ezh2 to Suz12 and reduce PRC2 chromatin engagement. Thus, in addition to serving as a histone chaperone and transcription elongation factor, Spt6 counteracts repression by opposing H3K27me3 deposition at critical genomic regulatory regions.
- Published
- 2017
19. gata2is Required for the runx1-Independent Hematopoiesis in Zebrafish
- Author
-
Erika M Kwon Kim, Vittorio Sartorelli, Paul P. Liu, Erica Bresciani, Stefania Dell'Orso, Kai Yu, Blake Carrington, Kevin Bishop, and Raman Sood
- Subjects
biology ,Immunology ,Cell Biology ,Hematology ,biology.organism_classification ,Biochemistry ,Phenotype ,Cell biology ,Loss of function mutation ,chemistry.chemical_compound ,Haematopoiesis ,RUNX1 ,chemistry ,hemic and lymphatic diseases ,embryonic structures ,Zebrafish ,Transcription factor - Abstract
The current notion about how hematopoietic stem cells (HSCs) are generated identifies the transcription factor RUNX1 as an essential factor for the emergence of definitive hematopoietic stem cells (HSCs) from the hemogenic endothelium. Consequently, Runx1knockout mice fail to develop definitive hematopoiesis and lack all definitive blood lineages and cannot survive past embryonic day 12. However, even though zebrafish with arunx1stop codon mutation (runx1W84X/W84X) presented defects in definitive hematopoiesis during embryogenesis, runx1W84X/W84Xembryos could develop to fertile adults with blood cells of multi-lineages, raising the possibility that HSCs can emerge without RUNX1. In order to determine if a RUNX1-independent mechanism can support the generation of HSCs we have generated three new zebrafish runx1-/- with engineered deletions of the runx1gene using TALEN and CRISPR-Cas9. Our analysis shows that all three mutants have identical phenotypei.e., failure to develop definitive hematopoiesis during early embryogenesis, with later reemergence of hematopoietic cells and survivalof therunx1 mutants to adulthood, further confirming the existence of a RUNX1-independent mechanism for the emergence of HSCs. In the absence of a functional runx1, a cd41-GFP+population of hematopoietic precursors can still be detected in the aorta-gonad-mesonephros (AGM) region and in the hematopoietic tissues of the mutant embryos. Single cell RNA sequencing of the wild type and mutant HSC/HSPC at embryonic and larval stages confirmed the presence of a population of runx1- /-cd41:GFPlow cells expressing HSC signature genes at 2.5 days post fertilization. At larval stages the runx1-/-HSCs maintain their ability to generate erythroid and myeloid lineage progenitors but they present a different expression profile compared to the wild type. In order to uncover the compensatory mechanism that drives the repopulation of the hematopoietic compartment in the absence of runx1we identified the molecular signatures that separate the runx1-/-HSC/HSPCs from the wild type and subsequently focused our attention on the transcription factors differentially expressed in the runx1-/-HSC/HSPCs. Our analysis shows that the master transcription factor gata2b is strongly upregulated in the runx1- /-HSCs during the recovery of hematopoiesis and it is also upregulated in the kidney marrow of the surviving runx1-/-adults. Given the key role of GATA2 in the HSC development and maintenance in both mouse and zebrafish, gata2b represented a strong candidate gene with the potential ability to drive the rescue of the runx1-/-phenotype. Indeed, a loss of function mutation or knock-down of gata2b can significantly reduce or abolish the survivability of the runx1-/-fish, indicating that gata2bis responsible for rescuing hematopoiesis in the runx1 mutant fish. Overall our results show that even though runx1 is necessary for the normal emergence of definitive HSCs in the embryos, in the absence of runx1the transcription factor gata2 is able to support definitive hematopoiesis that is sufficient for the embryos to develop to functional adults in the zebrafish. The current notion about how hematopoietic stem cells (HSCs) are generated identifies the transcription factor RUNX1 as an essential factor for the emergence of definitive hematopoietic stem cells (HSCs) from the hemogenic endothelium. Consequently, Runx1knockout mice fail to develop definitive hematopoiesis and lack all definitive blood lineages and cannot survive past embryonic day 12. However, even though zebrafish with arunx1stop codon mutation (runx1W84X/W84X) presented defects in definitive hematopoiesis during embryogenesis, runx1W84X/W84Xembryos could develop to fertile adults with blood cells of multi-lineages, raising the possibility that HSCs can emerge without RUNX1. In order to determine if a RUNX1-independent mechanism can support the generation of HSCs we have generated three new zebrafish runx1-/- with engineered deletions of the runx1gene using TALEN and CRISPR-Cas9. Our analysis shows that all three mutants have identical phenotypei.e., failure to develop definitive hematopoiesis during early embryogenesis, with later reemergence of hematopoietic cells and survivalof therunx1 mutants to adulthood, further confirming the existence of a RUNX1-independent mechanism for the emergence of HSCs. In the absence of a functional runx1, a cd41-GFP+population of hematopoietic precursors can still be detected in the aorta-gonad-mesonephros (AGM) region and in the hematopoietic tissues of the mutant embryos. Single cell RNA sequencing of the wild type and mutant HSC/HSPC at embryonic and larval stages confirmed the presence of a population of runx1- /-cd41:GFPlow cells expressing HSC signature genes at 2.5 days post fertilization. At larval stages the runx1-/-HSCs maintain their ability to generate erythroid and myeloid lineage progenitors but they present a different expression profile compared to the wild type. In order to uncover the compensatory mechanism that drives the repopulation of the hematopoietic compartment in the absence of runx1we identified the molecular signatures that separate the runx1-/-HSC/HSPCs from the wild type and subsequently focused our attention on the transcription factors differentially expressed in the runx1-/-HSC/HSPCs. Our analysis shows that the master transcription factor gata2b is strongly upregulated in the runx1- /-HSCs during the recovery of hematopoiesis and it is also upregulated in the kidney marrow of the surviving runx1-/-adults. Given the key role of GATA2 in the HSC development and maintenance in both mouse and zebrafish, gata2b represented a strong candidate gene with the potential ability to drive the rescue of the runx1-/-phenotype. Indeed, a loss of function mutation or knock-down of gata2b can significantly reduce or abolish the survivability of the runx1-/-fish, indicating that gata2bis responsible for rescuing hematopoiesis in the runx1 mutant fish. Overall our results show that even though runx1 is necessary for the normal emergence of definitive HSCs in the embryos, in the absence of runx1the transcription factor gata2 is able to support definitive hematopoiesis that is sufficient for the embryos to develop to functional adults in the zebrafish. Disclosures No relevant conflicts of interest to declare.
- Published
- 2019
- Full Text
- View/download PDF
20. Essential Role of SIRT1 Signaling in the Nucleus Accumbens in Cocaine and Morphine Action
- Author
-
Vittorio Sartorelli, Mitra Heshmati, Elizabeth A. Heller, Ja Wook Koo, Eric J. Nestler, Jian Feng, Rachael L. Neve, Jacqui Rabkin, Deveroux Ferguson, Xiaochuan Liu, Ning-Yi Shao, Li Shen, and William Renthal
- Subjects
Male ,Narcotics ,Chromatin Immunoprecipitation ,Dendritic spine ,Immunoblotting ,Drug action ,Nucleus accumbens ,Medium spiny neuron ,SIRT2 ,Nucleus Accumbens ,Cocaine-Related Disorders ,Mice ,Sirtuin 2 ,Cocaine ,Dopamine Uptake Inhibitors ,Reward ,Sirtuin 1 ,Animals ,Oligonucleotide Array Sequence Analysis ,Morphine ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,General Neuroscience ,Articles ,Mice, Inbred C57BL ,enzymes and coenzymes (carbohydrates) ,Sirtuin ,biology.protein ,Brain stimulation reward ,Morphine Dependence ,Neuroscience ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction - Abstract
Sirtuins (SIRTs), class III histone deacetylases, are well characterized for their control of cellular physiology in peripheral tissues, but their influence in brain under normal and pathological conditions remains poorly understood. Here, we establish an essential role for SIRT1 and SIRT2 in regulating behavioral responses to cocaine and morphine through actions in the nucleus accumbens (NAc), a key brain reward region. We show that chronic cocaine administration increases SIRT1 and SIRT2 expression in the mouse NAc, while chronic morphine administration induces SIRT1 expression alone, with no regulation of all other sirtuin family members observed. Drug induction of SIRT1 and SIRT2 is mediated in part at the transcriptional level via the drug-induced transcription factor ΔFosB and is associated with robust histone modifications at theSirt1andSirt2genes. Viral-mediated overexpression of SIRT1 or SIRT2 in the NAc enhances the rewarding effects of both cocaine and morphine. In contrast, the local knockdown of SIRT1 from the NAc of floxedSirt1mice decreases drug reward. Such behavioral effects of SIRT1 occur in concert with its regulation of numerous synaptic proteins in NAc as well as with SIRT1-mediated induction of dendritic spines on NAc medium spiny neurons. These studies establish sirtuins as key mediators of the molecular and cellular plasticity induced by drugs of abuse in NAc, and of the associated behavioral adaptations, and point toward novel signaling pathways involved in drug action.
- Published
- 2013
- Full Text
- View/download PDF
21. The methyltransferase SMYD3 mediates the recruitment of transcriptional cofactors at the myostatin and c-Met genes and regulates skeletal muscle atrophy
- Author
-
James G. Ryall, Vittorio Sartorelli, Raffaella Fittipaldi, Valentina Proserpio, and Giuseppina Caretti
- Subjects
muscle atrophy ,Transcription, Genetic ,Positive Transcriptional Elongation Factor B ,Muscle Fibers, Skeletal ,Muscle Proteins ,Myostatin ,Dexamethasone ,Cell Line ,Mice ,Phosphoserine ,Atrophy ,SMYD3 ,BRD4 ,myostatin ,Genetics ,medicine ,Transcriptional regulation ,Animals ,Phosphorylation ,Muscle, Skeletal ,Transcription factor ,SKP Cullin F-Box Protein Ligases ,biology ,Nuclear Proteins ,Skeletal muscle ,Histone-Lysine N-Methyltransferase ,Proto-Oncogene Proteins c-met ,medicine.disease ,Cyclin-Dependent Kinase 9 ,Muscle atrophy ,Muscular Atrophy ,medicine.anatomical_structure ,GDF11 ,Cancer research ,biology.protein ,RNA Polymerase II ,medicine.symptom ,Protein Binding ,Transcription Factors ,Research Paper ,Developmental Biology - Abstract
Elucidating the epigenetic mechanisms underlying muscle mass determination and skeletal muscle wasting holds the potential of identifying molecular pathways that constitute possible drug targets. Here, we report that the methyltransferase SMYD3 modulates myostatin and c-Met transcription in primary skeletal muscle cells and C2C12 myogenic cells. SMYD3 targets the myostatin and c-Met genes and participates in the recruitment of the bromodomain protein BRD4 to their regulatory regions through protein–protein interaction. By recruiting BRD4, SMYD3 favors chromatin engagement of the pause–release factor p-TEFb (positive transcription elongation factor) and elongation of Ser2-phosphorylated RNA polymerase II (PolIISer2P). Reducing SMYD3 decreases myostatin and c-Met transcription, thus protecting from glucocorticoid-induced myotube atrophy. Supporting functional relevance of the SMYD3/BRD4 interaction, BRD4 pharmacological blockade by the small molecule JQ1 prevents dexamethasone-induced myostatin and atrogene up-regulation and spares myotube atrophy. Importantly, in a mouse model of dexamethasone-induced skeletal muscle atrophy, SMYD3 depletion prevents muscle loss and fiber size decrease. These findings reveal a mechanistic link between SMYD3/BRD4-dependent transcriptional regulation, muscle mass determination, and skeletal muscle atrophy and further encourage testing of small molecules targeting specific epigenetic regulators in animal models of muscle wasting.
- Published
- 2013
- Full Text
- View/download PDF
22. The histone chaperone Spt6 coordinates histone H3K27 demethylation and myogenesis
- Author
-
A. Hongjun Wang, Chaochen Wang, Vittorio Sartorelli, Cara E. Moravec, Hossein Zare, Gustavo Gutierrez-Cruz, Kai Ge, Kambiz Mousavi, and Howard I. Sirotkin
- Subjects
General Immunology and Microbiology ,General Neuroscience ,Cellular differentiation ,macromolecular substances ,Biology ,MyoD ,Molecular biology ,General Biochemistry, Genetics and Molecular Biology ,Chromatin ,Histone methyltransferase ,Histone H2A ,biology.protein ,Histone code ,PRC2 ,Molecular Biology ,Chromatin immunoprecipitation - Abstract
Histone chaperones affect chromatin structure and gene expression through interaction with histones and RNA polymerase II (PolII). Here, we report that the histone chaperone Spt6 counteracts H3K27me3, an epigenetic mark deposited by the Polycomb Repressive Complex 2 (PRC2) and associated with transcriptional repression. By regulating proper engagement and function of the H3K27 demethylase KDM6A (UTX), Spt6 effectively promotes H3K27 demethylation, muscle gene expression, and cell differentiation. ChIP-Seq experiments reveal an extensive genome-wide overlap of Spt6, PolII, and KDM6A at transcribed regions that are devoid of H3K27me3. Mammalian cells and zebrafish embryos with reduced Spt6 display increased H3K27me3 and diminished expression of the master regulator MyoD, resulting in myogenic differentiation defects. As a confirmation for an antagonistic relationship between Spt6 and H3K27me3, inhibition of PRC2 permits MyoD re-expression in myogenic cells with reduced Spt6. Our data indicate that, through cooperation with PolII and KDM6A, Spt6 orchestrates removal of H3K27me3, thus controlling developmental gene expression and cell differentiation.
- Published
- 2013
- Full Text
- View/download PDF
23. Specific Sirt1 Activator-mediated Improvement in Glucose Homeostasis Requires Sirt1-Independent Activation of AMPK
- Author
-
James G. Ryall, Alexandra L. Brown, Jay H. Chung, Hyeog Kang, Myung K. Kim, Jee Hyun Um, Vittorio Sartorelli, Andrew Philp, Hengming Ke, Xihui Xu, Sung Jun Park, Xuesong Feng, Faiyaz Ahmad, and Simon Schenk
- Subjects
AMPK ,0301 basic medicine ,Aging ,Epac ,lcsh:Medicine ,SRT1720 ,Mitochondrion ,AMP-Activated Protein Kinases ,Mice ,Sirtuin 1 ,Cyclic AMP ,Glucose homeostasis ,Guanine Nucleotide Exchange Factors ,Phosphorylation ,RNA, Small Interfering ,Mice, Knockout ,Sirt1 ,lcsh:R5-920 ,Chemistry ,food and beverages ,Phosphodiesterase ,Type 2 diabetes ,General Medicine ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,3. Good health ,Cell biology ,Mitochondria ,Phosphodiesterases ,Knockout mouse ,RNA Interference ,lcsh:Medicine (General) ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction ,Heterocyclic Compounds, 4 or More Rings ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,cAMP ,Glucose Intolerance ,Animals ,Humans ,Obesity ,Protein kinase A ,Activator (genetics) ,Phosphoric Diester Hydrolases ,lcsh:R ,Mice, Inbred C57BL ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,Glucose ,Mutagenesis, Site-Directed ,Commentary ,HeLa Cells - Abstract
The specific Sirt1 activator SRT1720 increases mitochondrial function in skeletal muscle, presumably by activating Sirt1. However, Sirt1 gain of function does not increase mitochondrial function, which raises a question about the central role of Sirt1 in SRT1720 action. Moreover, it is believed that the metabolic effects of SRT1720 occur independently of AMP-activated protein kinase (AMPK), an important metabolic regulator that increases mitochondrial function. Here, we show that SRT1720 activates AMPK in a Sirt1-independent manner and SRT1720 activates AMPK by inhibiting a cAMP degrading phosphodiesterase (PDE) in a competitive manner. Inhibiting the cAMP effector protein Epac prevents SRT1720 from activating AMPK or Sirt1 in myotubes. Moreover, SRT1720 does not increase mitochondrial function or improve glucose tolerance in AMPKα2 knockout mice. Interestingly, weight loss induced by SRT1720 is not sufficient to improve glucose tolerance. Therefore, contrary to current belief, the metabolic effects produced by SRT1720 require AMPK, which can be activated independently of Sirt1.
- Published
- 2017
24. Integrated expression analysis of muscle hypertrophy identifies Asb2 as a negative regulator of muscle mass
- Author
-
Louise Cunningham, Marco Sandri, David E. James, Paul Gregorevic, Hongwei Qian, Benjamin L. Parker, Jonathan R. Davey, James G. Ryall, Kevin I. Watt, Rima Chaudhuri, Jeffrey S. Chamberlain, and Vittorio Sartorelli
- Subjects
0301 basic medicine ,biology ,business.industry ,Regulator ,Physiology ,Skeletal muscle ,General Medicine ,Myostatin ,medicine.disease ,Muscle hypertrophy ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Sarcopenia ,biology.protein ,medicine ,business ,Psychological repression ,Follistatin ,Transforming growth factor - Abstract
The transforming growth factor-β (TGF-β) signaling network is a critical regulator of skeletal muscle mass and function and, thus, is an attractive therapeutic target for combating muscle disease, but the underlying mechanisms of action remain undetermined. We report that follistatin-based interventions (which modulate TGF-β network activity) can promote muscle hypertrophy that ameliorates aging-associated muscle wasting. However, the muscles of old sarcopenic mice demonstrate reduced response to follistatin compared with healthy young-adult musculature. Quantitative proteomic and transcriptomic analyses of young-adult muscles identified a transcription/translation signature elicited by follistatin exposure, which included repression of ankyrin repeat and SOCS box protein 2 (Asb2). Increasing expression of ASB2 reduced muscle mass, thereby demonstrating that Asb2 is a TGF-β network-responsive negative regulator of muscle mass. In contrast to young-adult muscles, sarcopenic muscles do not exhibit reduced ASB2 abundance with follistatin exposure. Moreover, preventing repression of ASB2 in young-adult muscles diminished follistatin-induced muscle hypertrophy. These findings provide insight into the program of transcription and translation events governing follistatin-mediated adaptation of skeletal muscle attributes and identify Asb2 as a regulator of muscle mass implicated in the potential mechanistic dysfunction between follistatin-mediated muscle growth in young and old muscles.
- Published
- 2016
- Full Text
- View/download PDF
25. Laminopathies disrupt epigenomic developmental programs and cell fate
- Author
-
Vittorio Sartorelli, Corinne Vigouroux, Kamel Mamchaoui, Stefania Dell'Orso, Gisèle Bonne, Eric P. Hoffman, Viola F. Gnochi, Jyoti K. Jaiswal, Vincent Mouly, and Jelena Perovanovic
- Subjects
0301 basic medicine ,Chromatin Immunoprecipitation ,Site-Specific DNA-Methyltransferase (Adenine-Specific) ,Satellite Cells, Skeletal Muscle ,Euchromatin ,Heterochromatin ,Muscle Fibers, Skeletal ,Mutation, Missense ,Emerin ,In Vitro Techniques ,Gene mutation ,Biology ,Real-Time Polymerase Chain Reaction ,Article ,Epigenesis, Genetic ,Myoblasts ,LMNA ,Mice ,03 medical and health sciences ,Animals ,Humans ,Missense mutation ,Cells, Cultured ,Cell Nucleus ,Genetics ,SOXB1 Transcription Factors ,Cell Cycle ,Computational Biology ,Cell Differentiation ,General Medicine ,DNA Methylation ,Lamin Type A ,Muscular Dystrophy, Emery-Dreifuss ,Chromatin ,HEK293 Cells ,030104 developmental biology ,Mutation ,embryonic structures ,Lamin ,Protein Binding - Abstract
The nuclear envelope protein lamin A is encoded by the lamin A/C (LMNA) gene, which can contain missense mutations that cause Emery-Dreifuss muscular dystrophy (EDMD) (p.R453W). We fused mutated forms of the lamin A protein to bacterial DNA adenine methyltransferase (Dam) to define euchromatic-heterochromatin (epigenomic) transitions at the nuclear envelope during myogenesis (using DamID-seq). Lamin A missense mutations disrupted appropriate formation of lamin A–associated heterochromatin domains in an allele-specific manner—findings that were confirmed by chromatin immunoprecipitation–DNA sequencing (ChIP-seq) in murine H2K cells and DNA methylation studies in fibroblasts from muscular dystrophy patient who carried a distinct LMNA mutation (p.H222P). Observed perturbations of the epigenomic transitions included exit from pluripotency and cell cycle programs [euchromatin (open, transcribed) to heterochromatin (closed, silent)], as well as induction of myogenic loci (heterochromatin to euchromatin). In muscle biopsies from patients with either a gain- or change-of-function LMNA gene mutation or a loss-of-function mutation in the emerin gene, both of which cause EDMD, we observed inappropriate loss of heterochromatin formation at the Sox2 pluripotency locus, which was associated with persistent mRNA expression of Sox2. Overexpression of Sox2 inhibited myogenic differentiation in human immortalized myoblasts. Our findings suggest that nuclear envelopathies are disorders of developmental epigenetic programming that result from altered formation of lamina-associated domains.
- Published
- 2016
- Full Text
- View/download PDF
26. Lysine methyltransferase G9a methylates the transcription factor MyoD and regulates skeletal muscle differentiation
- Author
-
Martin J. Walsh, Vittorio Sartorelli, Teng-Kai Chung, Reshma Taneja, Yong Hua Tan, Suma Gopinadhan, Belinda Mei Tze Ling, SiDe Li, Wai Kay Kok, Narendra Bharathy, and Vinay Kumar Rao
- Subjects
Cellular differentiation ,Molecular Sequence Data ,Biology ,Muscle Development ,MyoD ,Methylation ,Cell Line ,Mice ,Histone H3 ,MyoD Protein ,Animals ,Humans ,Amino Acid Sequence ,Muscle, Skeletal ,Myogenin ,Multidisciplinary ,PITX2 ,Myogenesis ,Lysine ,Cell Differentiation ,Histone-Lysine N-Methyltransferase ,Biological Sciences ,Biochemistry ,Myogenic regulatory factors ,Protein Binding - Abstract
Skeletal muscle cells have served as a paradigm for understanding mechanisms leading to cellular differentiation. The proliferation and differentiation of muscle precursor cells require the concerted activity of myogenic regulatory factors including MyoD. In addition, chromatin modifiers mediate dynamic modifications of histone tails that are vital to reprogramming cells toward terminal differentiation. Here, we provide evidence for a unique dimension to epigenetic regulation of skeletal myogenesis. We demonstrate that the lysine methyltransferase G9a is dynamically expressed in myoblasts and impedes differentiation in a methyltransferase activity-dependent manner. In addition to mediating histone H3 lysine-9 di-methylation (H3K9me2) on MyoD target promoters, endogenous G9a interacts with MyoD in precursor cells and directly methylates it at lysine 104 (K104) to constrain its transcriptional activity. Mutation of K104 renders MyoD refractory to inhibition by G9a and enhances its myogenic activity. Interestingly, MyoD methylation is critical for G9a-mediated inhibition of myogenesis. These findings provide evidence of an unanticipated role for methyltransferases in cellular differentiation states by direct posttranslational modification of a transcription factor.
- Published
- 2012
- Full Text
- View/download PDF
27. A Muscle-Specific Enhancer RNA Mediates Cohesin Recruitment and Regulates Transcription In trans
- Author
-
Daniel R. Larson, Jelena Perovanovic, Vittorio Sartorelli, Kyung-Dae Ko, Joseph Rodriguez, Laura Vian, Pei-Fang Tsai, Thomas Ried, Markus Hafner, A. Hongjun Wang, Michelle D Tran, Aster H. Juan, Hong-Wei Sun, Karinna O. Vivanco, Darawalee Wangsa, Kan Jiang, Stefania Dell'Orso, Dimitrios G. Anastasakis, Evelyn Ralston, and Aishe A. Sarshad
- Subjects
0301 basic medicine ,RNA, Untranslated ,Transcription, Genetic ,Cohesin complex ,Chromosomal Proteins, Non-Histone ,Cell Cycle Proteins ,Enhancer RNAs ,Biology ,MyoD ,Mice ,03 medical and health sciences ,Animals ,Humans ,Muscle, Skeletal ,Enhancer ,Molecular Biology ,Myogenin ,MyoD Protein ,Cohesin loading ,Cohesin ,Muscle cell differentiation ,Cell Differentiation ,Cell Biology ,musculoskeletal system ,Chromatin ,Cell biology ,Enhancer Elements, Genetic ,HEK293 Cells ,030104 developmental biology ,biological phenomena, cell phenomena, and immunity ,tissues - Abstract
The enhancer regions of the myogenic master regulator MyoD give rise to at least two enhancer RNAs. Core enhancer eRNA (CEeRNA) regulates transcription of the adjacent MyoD gene, whereas DRReRNA affects expression of Myogenin in trans. We found that DRReRNA is recruited at the Myogenin locus, where it colocalizes with Myogenin nascent transcripts. DRReRNA associates with the cohesin complex, and this association correlates with its transactivating properties. Despite being expressed in undifferentiated cells, cohesin is not loaded on Myogenin until the cells start expressing DRReRNA, which is then required for cohesin chromatin recruitment and maintenance. Functionally, depletion of either cohesin or DRReRNA reduces chromatin accessibility, prevents Myogenin activation, and hinders muscle cell differentiation. Thus, DRReRNA ensures spatially appropriate cohesin loading in trans to regulate gene expression.
- Published
- 2018
- Full Text
- View/download PDF
28. SirT1 in muscle physiology and disease: lessons from mouse models
- Author
-
Vittorio Sartorelli, Nadia Rosenthal, Andreas G. Ladurner, Marcella Fulco, and Manlio Vinciguerra
- Subjects
Transgene ,Calorie restriction ,Circadian clock ,ved/biology.organism_classification_rank.species ,Neuroscience (miscellaneous) ,Medicine (miscellaneous) ,Physiology ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Sirtuin 1 ,Immunology and Microbiology (miscellaneous) ,medicine ,Animals ,Humans ,Disease ,Model organism ,biology ,ved/biology ,Muscles ,Skeletal muscle ,Cell biology ,Disease Models, Animal ,medicine.anatomical_structure ,Perspective ,Sirtuin ,biology.protein ,NAD+ kinase - Abstract
Sirtuin 1 (SirT1) is the largest of the seven members of the sirtuin family of class III nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylases, whose activation is beneficial for metabolic, neurodegenerative, inflammatory and neoplastic diseases, and augments life span in model organisms (Finkel et al., 2009; Lavu et al., 2008). In vitro studies show that SirT1 protects genome integrity and is involved in circadian physiological rhythms (Asher et al., 2008; Nakahata et al., 2008; Oberdoerffer et al., 2008). In the last few years, a fundamental role for SirT1 in the metabolism and differentiation of skeletal muscle cells has been uncovered (Fulco et al., 2003), and the use of specific transgenic or knockout SirT1 mouse models implicates it in the protection of heart muscle from oxidative and hypertrophic stresses (Alcendor et al., 2007). In this Perspective, we review the recent exciting findings that have established a key role for the ’longevity’ protein SirT1 in skeletal and heart muscle physiology and disease. Furthermore, given the multiple biological functions of SirT1, we discuss the unique opportunities that SirT1 mouse models can offer to improve our integrated understanding of the metabolism, as well as the regeneration and aging-associated changes in the circadian function, of skeletal and heart muscle.
- Published
- 2010
- Full Text
- View/download PDF
29. Glucose Restriction Inhibits Skeletal Myoblast Differentiation by Activating SIRT1 through AMPK-Mediated Regulation of Nampt
- Author
-
Yana Cen, Marcella Fulco, Vittorio Sartorelli, Po Zhao, Anthony A. Sauve, Michael W. McBurney, and Eric P. Hoffman
- Subjects
Transcription, Genetic ,Myoblasts, Skeletal ,HUMDISEASE ,Nicotinamide phosphoribosyltransferase ,DEVBIO ,AMP-Activated Protein Kinases ,Protein Serine-Threonine Kinases ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Mice ,chemistry.chemical_compound ,Sirtuin 1 ,AMP-activated protein kinase ,Multienzyme Complexes ,medicine ,Animals ,Sirtuins ,Myocyte ,Nicotinamide Phosphoribosyltransferase ,Molecular Biology ,Mice, Inbred BALB C ,biology ,Myogenesis ,AMPK ,Skeletal muscle ,Cell Differentiation ,Cell Biology ,NAD ,Enzyme Activation ,Glucose ,medicine.anatomical_structure ,Gene Expression Regulation ,chemistry ,Biochemistry ,biology.protein ,Cytokines ,NAD+ kinase ,Food Deprivation ,Developmental Biology - Abstract
SummaryIt is intuitive to speculate that nutrient availability may influence differentiation of mammalian cells. Nonetheless, a comprehensive complement of the molecular determinants involved in this process has not been elucidated yet. Here, we have investigated how nutrients (glucose) affect skeletal myogenesis. Glucose restriction (GR) impaired differentiation of skeletal myoblasts and was associated with activation of the AMP-activated protein kinase (AMPK). Activated AMPK was required to promote GR-induced transcription of the NAD+ biosynthetic enzyme Nampt. Indeed, GR augmented the Nampt activity, which consequently modified the intracellular [NAD+]:[NADH] ratio and nicotinamide levels, and mediated inhibition of skeletal myogenesis. Skeletal myoblasts derived from SIRT1+/− heterozygous mice were resistant to the effects of either GR or AMPK activation. These experiments reveal that AMPK, Nampt, and SIRT1 are the molecular components of a functional signaling pathway that allows skeletal muscle cells to sense and react to nutrient availability.
- Published
- 2008
- Full Text
- View/download PDF
30. p68 (Ddx5) interacts with Runx2 and regulates osteoblast differentiation
- Author
-
Vittorio Sartorelli, Gary S. Stein, Lingling Niu, Giuseppina Caretti, Andre J. Van Wijnen, Jennifer J. Westendorf, Eric D. Jensen, Frances V. Fuller-Pace, Samantha M. Nicol, and Nadiya M. Teplyuk
- Subjects
musculoskeletal diseases ,Core Binding Factor Alpha 1 Subunit ,Biology ,Biochemistry ,DEAD-box RNA Helicases ,Mice ,chemistry.chemical_compound ,stomatognathic system ,Transcription (biology) ,Chlorocebus aethiops ,Gene expression ,medicine ,Animals ,Molecular Biology ,Transcription factor ,Cell Nucleus ,Osteoblasts ,DDX5 ,Stem Cells ,musculoskeletal, neural, and ocular physiology ,Skull ,Cell Differentiation ,Osteoblast ,Cell Biology ,musculoskeletal system ,RNA Helicase A ,Molecular biology ,Mice, Mutant Strains ,RUNX2 ,medicine.anatomical_structure ,Gene Expression Regulation ,chemistry ,COS Cells ,embryonic structures ,Mesodermal cell differentiation ,Protein Binding - Abstract
Runx2 is an essential transcription factor for osteoblast development from mesenchymal progenitors. Runx2 regulates gene expression by interacting with numerous transcription factors and co-activators to integrate signaling events within the nucleus. In this study we used affinity purification and proteomic techniques to identify novel Runx2 interacting proteins. One of these proteins is the DEAD box RNA helicase, p68 (Ddx5). p68 regulates many aspects of RNA expression, including transcription and splicing. p68 co-localized with Runx2 in punctate foci within the nucleus. In transcription assays, p68 functioned as a co-activator of Runx2, but its helicase activity was not essential for co-activation. In accordance, Runx2 transcriptional activity was muted in p68-suppressed cells. Surprisingly, osteoblast differentiation of the multipotent progenitor C2C12 cell line was accelerated by p68 suppression and Runx2 suppressed p68 expression in calvarial progenitor cells. Together these data demonstrate that p68 is a novel co-activator for Runx2, but it inhibits osteogenic differentiation of progenitor cells. Moreover Runx2 has an active role in regulating p68 levels in osteoblast precursors. Thus, crosstalk between Runx2 and p68 controls osteoblast specification and maturation at multiple levels.
- Published
- 2008
- Full Text
- View/download PDF
31. Polycomb Ezh2 controls the fate of GABAergic neurons in the embryonic cerebellum
- Author
-
Vittorio Sartorelli, Aster H. Juan, Hossein Zare, Hongjun A. Wang, Kyung Dae Ko, and Xuesong Feng
- Subjects
0301 basic medicine ,Cell type ,Cerebellum ,Transcription, Genetic ,Cell Count ,macromolecular substances ,Biology ,Bioinformatics ,Methylation ,Histones ,Purkinje Cells ,03 medical and health sciences ,Interneurons ,medicine ,Animals ,Cell Lineage ,Enhancer of Zeste Homolog 2 Protein ,Epigenetics ,GABAergic Neurons ,Molecular Biology ,Transcription factor ,Rhombic lip ,Cell Proliferation ,Mice, Knockout ,Genome ,Lysine ,Tumor Suppressor Proteins ,Neurogenesis ,EZH2 ,Gene Expression Regulation, Developmental ,PAX7 Transcription Factor ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,Genetic Loci ,GABAergic ,Gene Deletion ,Research Article ,Developmental Biology - Abstract
While the genetic interactions between signaling pathways and transcription factors have been largely decoded, much remains to be learned about the epigenetic regulation of cerebellar development. Here, we report that cerebellar deletion of Ezh2, the methyltransferase subunit of the PRC2 complex, results in reduced H3K27me3 and profound transcriptional dysregulation, including that of a set of transcription factors directly involved in cerebellar neuronal cell type specification and differentiation. Such transcriptional changes led to increased GABAergic interneurons and decreased.Purkinje cells. Transcriptional changes also inhibited the proliferation of granule precursor cells derived from the rhombic lip. The loss of both cell types ultimately resulted in cerebellar hypoplasia. These findings indicate Ezh2/PRC2 plays critical roles in regulating neurogenesis from both cerebellar germinal zones.
- Published
- 2016
- Full Text
- View/download PDF
32. MyoD Acetylation Influences Temporal Patterns of Skeletal Muscle Gene Expression
- Author
-
Eric P. Hoffman, Giuseppina Caretti, Vittorio Sartorelli, Po Zhao, and Monica Di Padova
- Subjects
Time Factors ,animal structures ,Transcription, Genetic ,Mice, Transgenic ,RNA polymerase II ,MyoD ,Biochemistry ,Mice ,Animals ,Cell Lineage ,Muscle, Skeletal ,Molecular Biology ,Myogenin ,MyoD Protein ,Genome ,Models, Genetic ,biology ,PITX2 ,Myogenesis ,Gene Expression Regulation, Developmental ,Cell Biology ,Fibroblasts ,musculoskeletal system ,Molecular biology ,Chromatin ,Histone ,biology.protein ,RNA Polymerase II ,Protein Processing, Post-Translational ,tissues ,Chromatin immunoprecipitation - Abstract
MyoD is sufficient to initiate the skeletal muscle gene expression program. Transcription of certain MyoD target genes occurs in the early phases, whereas that of others is induced only at later stages, although MyoD is present throughout the differentiation process. MyoD acetylation regulates transcriptional competency, yet whether this post-translational modification is equally relevant for activation of all the MyoD targets is unknown. Moreover, the molecular mechanisms through which acetylation ensures that MyoD achieves its optimal activity remain unexplored. To address these two outstanding issues, we have coupled genome-wide expression profiling and chromatin immunoprecipitation in a model system in which MyoD or its nonacetylatable version was inducibly activated in mouse embryonic fibroblasts derived from MyoD(-/-)/Myf5(-/-) mice. Our results reveal that MyoD acetylation influences transcription of selected genes expressed at defined stages of the muscle program by regulating chromatin access of MyoD, histone acetylation, and RNA polymerase II recruitment.
- Published
- 2007
- Full Text
- View/download PDF
33. EZH2 is crucial for both differentiation of regulatory T cells and T effector cell expansion
- Author
-
Yuka Kanno, Hong-Wei Sun, Michael Bonelli, Vittorio Sartorelli, Golnaz Vahedi, Arian Laurence, Kan Jiang, Kiyoshi Hirahara, Giuseppe Sciumè, Xiang-Ping Yang, John J. O'Shea, Behdad Afzali, and Dragana Jankovic
- Subjects
Knockout ,T-Lymphocytes ,chemical and pharmacologic phenomena ,macromolecular substances ,Biology ,T-Lymphocytes, Regulatory ,Article ,Autoimmune Diseases ,Mice ,Interleukin 21 ,Animals ,Cytotoxic T cell ,Enhancer of Zeste Homolog 2 Protein ,IL-2 receptor ,Antigen-presenting cell ,Mice, Knockout ,Cell Differentiation ,Colitis ,Cytokines ,Gene Expression Regulation ,Polycomb Repressive Complex 2 ,Multidisciplinary ,CD40 ,ZAP70 ,FOXP3 ,hemic and immune systems ,Natural killer T cell ,Regulatory ,Cell biology ,Immunology ,biology.protein - Abstract
The roles of EZH2 in various subsets of CD4+ T cells are controversial and its mechanisms of action are incompletely understood. FOXP3-positive Treg cells are a critical helper T cell subset and dysregulation of Treg generation or function results in systemic autoimmunity. FOXP3 associates with EZH2 to mediate gene repression and suppressive function. Herein, we demonstrate that deletion of Ezh2 in CD4 T cells resulted in reduced numbers of Treg cells in vivo and differentiation in vitro and an increased proportion of memory CD4 T cells in part due to exaggerated production of effector cytokines. Furthermore, we found that both Ezh2-deficient Treg cells and T effector cells were functionally impaired in vivo: Tregs failed to constrain autoimmune colitis and T effector cells neither provided a protective response to T. gondii infection nor mediated autoimmune colitis. The dichotomous function of EZH2 in regulating differentiation and senescence in effector and regulatory T cells helps to explain the apparent existing contradictions in literature.
- Published
- 2015
- Full Text
- View/download PDF
34. The Histone Variant MacroH2A1.2 Is Necessary for the Activation of Muscle Enhancers and Recruitment of the Transcription Factor Pbx1
- Author
-
Han-Yu Shih, Vittorio Sartorelli, Libera Berghella, A. Hongjun Wang, Gustavo Gutierrez-Cruz, Kayoko Saso, Andreas G. Ladurner, Stefania Dell'Orso, John J. O'Shea, and Hossein Zare
- Subjects
0301 basic medicine ,Transcription, Genetic ,Enhancer RNAs ,SAP30 ,Muscle Development ,General Biochemistry, Genetics and Molecular Biology ,Article ,Epigenesis, Genetic ,Histones ,03 medical and health sciences ,Mice ,Histone H1 ,Histone H2A ,Histone code ,Animals ,Humans ,Gene Regulatory Networks ,Enhancer ,Muscle, Skeletal ,lcsh:QH301-705.5 ,Transcription factor ,MyoD Protein ,Genetics ,Homeodomain Proteins ,Muscle Cells ,Genome ,biology ,fungi ,Pre-B-Cell Leukemia Transcription Factor 1 ,Acetylation ,Cell Differentiation ,Chromatin ,030104 developmental biology ,Histone ,Enhancer Elements, Genetic ,HEK293 Cells ,lcsh:Biology (General) ,biology.protein ,Transcriptome ,Protein Binding ,Transcription Factors - Abstract
Summary: Histone variants complement and integrate histone post-translational modifications in regulating transcription. The histone variant macroH2A1 (mH2A1) is almost three times the size of its canonical H2A counterpart, due to the presence of an ∼25 kDa evolutionarily conserved non-histone macro domain. Strikingly, mH2A1 can mediate both gene repression and activation. However, the molecular determinants conferring these alternative functions remain elusive. Here, we report that mH2A1.2 is required for the activation of the myogenic gene regulatory network and muscle cell differentiation. H3K27 acetylation at prospective enhancers is exquisitely sensitive to mH2A1.2, indicating a role of mH2A1.2 in imparting enhancer activation. Both H3K27 acetylation and recruitment of the transcription factor Pbx1 at prospective enhancers are regulated by mH2A1.2. Overall, our findings indicate a role of mH2A1.2 in marking regulatory regions for activation. : Dell’Orso et al. report that the histone variant macroH2A1.2 is required for activation of muscle-gene expression and cell differentiation. Genome-wide analyses indicate that macroH2A1.2 is enriched at prospective muscle-specific enhancers where it is required for H3K27 acetylation and recruitment of the transcription factor Pbx1.
- Published
- 2015
35. Follistatin induction by nitric oxide through cyclic GMP: a tightly regulated signaling pathway that controls myoblast fusion
- Author
-
Silvia Baesso, Silvia Brunelli, Giulio Cossu, Monica Di Padova, Emilio Clementi, Clara De Palma, Addolorata Pisconti, Vittorio Sartorelli, Daniela Deponti, Pisconti, A, Brunelli, S, Di Padova, M, De Palma, C, Deponti, D, Baesso, S, Sartorelli, V, Cossu, G, and Clementi, E
- Subjects
Follistatin ,medicine.medical_specialty ,Transcription, Genetic ,Myoblasts, Skeletal ,Muscle Development ,Nitric Oxide ,Article ,Cell Fusion ,Mice ,chemistry.chemical_compound ,Myoblast fusion ,MyoD Protein ,Internal medicine ,medicine ,Animals ,follistatin, nitric oxide, satellite cells ,Cyclic AMP Response Element-Binding Protein ,Muscle, Skeletal ,Cyclic GMP ,Cyclic guanosine monophosphate ,Cells, Cultured ,Research Articles ,Cell fusion ,NFATC Transcription Factors ,biology ,Myogenesis ,BIO/13 - BIOLOGIA APPLICATA ,Cell Biology ,Retraction ,Cell biology ,Nitric oxide synthase ,Endocrinology ,chemistry ,biology.protein ,Female ,Nitric Oxide Synthase ,Signal transduction ,Signal Transduction - Abstract
The mechanism of skeletal myoblast fusion is not well understood. We show that endogenous nitric oxide (NO) generation is required for myoblast fusion both in embryonic myoblasts and in satellite cells. The effect of NO is concentration and time dependent, being evident only at the onset of differentiation, and direct on the fusion process itself. The action of NO is mediated through a tightly regulated activation of guanylate cyclase and generation of cyclic guanosine monophosphate (cGMP), so much so that deregulation of cGMP signaling leads to a fusion-induced hypertrophy of satellite-derived myotubes and embryonic muscles, and to the acquisition of fusion competence by myogenic precursors in the presomitic mesoderm. NO and cGMP induce expression of follistatin, and this secreted protein mediates their action in myogenesis. These results establish a hitherto unappreciated role of NO and cGMP in regulating myoblast fusion and elucidate their mechanism of action, providing a direct link with follistatin, which is a key player in myogenesis.
- Published
- 2006
- Full Text
- View/download PDF
36. Fgfr4 Is Required for Effective Muscle Regeneration in Vivo
- Author
-
Vittorio Sartorelli, Eric P. Hoffman, Stephanie J. Mitchell, Adele L. Boskey, Giuseppina Caretti, Wallace L. McKeehan, Lauren M. Pachman, and Po Zhao
- Subjects
MyoD Protein ,Myogenesis ,Cellular differentiation ,Myosin ,Cell Biology ,Biology ,MyoD ,TEAD2 ,Molecular Biology ,Biochemistry ,Molecular biology ,Chromatin immunoprecipitation ,Transcription factor - Abstract
Fgfr4 has been shown to be important for appropriate muscle development in chick limb buds; however, Fgfr4 null mice show no phenotype. Here, we show that staged induction of muscle regeneration in Fgfr4 null mice becomes highly abnormal at the time point when Fgfr4 is normally expressed. By 7 days of regeneration, differentiation of myotubes became poorly coordinated and delayed by both histology and embryonic myosin heavy chain staining. By 14 days much of the muscle was replaced by fat and calcifications. To begin to dissect the molecular pathways involving Fgfr4, we queried the promoter sequences for transcriptional factor binding sites and tested candidate regulators in a 27-time point regeneration series. The Fgfr4 promoter region contained a Tead protein binding site (M-CAT 5′-CATTCCT-3′), and Tead2 showed induction during regeneration commensurate with Fgfr4 regulation. Co-transfection of Tead2 and Fgfr4 promoter reporter constructs into C2C12 myotubes showed Tead2 to activate Fgfr4, and mutation of the M-CAT motif in the Fgfr4 promoter abolished these effects. Immunostaining for Tead2 showed timed expression in myotube nuclei consistent with the mRNA data. Query of the expression timing and genomic sequences of Tead2 suggested direct regulation by MyoD, and consistent with this, MyoD directly bound to two strong E-boxes in the first intron of Tead2 by chromatin immunoprecipitation assay. Moreover, co-transfection of MyoD and Tead2 intron reporter constructs into 10T1/2 cells activated reporter activity in a dose-dependent manner. This activation was greatly reduced when the two E-boxes were mutated. Our data suggest a novel MyoD-Tead2-Fgfr4 pathway important for effective muscle regeneration.
- Published
- 2006
- Full Text
- View/download PDF
37. Mechanisms underlying the transcriptional regulation of skeletal myogenesis
- Author
-
Giuseppina Caretti and Vittorio Sartorelli
- Subjects
Genetics ,Regulation of gene expression ,Transcription, Genetic ,Myogenesis ,Gene Expression Regulation, Developmental ,E-box ,TCF4 ,Biology ,Article ,Chromatin ,DNA-Binding Proteins ,Basic Helix-Loop-Helix Transcription Factors ,Transcriptional regulation ,Animals ,Humans ,E2F1 ,Muscle, Skeletal ,Enhancer ,Transcription Factors ,Developmental Biology - Abstract
During skeletal myogenesis, chromatin-modifying enzymes are engaged at discrete genomic regions by transcription factors that recognize sequence-specific DNA motifs located at muscle gene regulatory regions. The composition of the chromatin-bound protein complexes and their temporally and spatially regulated recruitment influence gene expression. Recent findings are consistent with the concept that chromatin modifiers play an important role in regulating skeletal muscle gene expression and cellular differentiation.
- Published
- 2005
- Full Text
- View/download PDF
38. The Polycomb Ezh2 methyltransferase regulates muscle gene expression and skeletal muscle differentiation
- Author
-
Vittorio Sartorelli, Gary E. Lyons, Monica Di Padova, Bruce Micales, and Giuseppina Caretti
- Subjects
Transcriptional Activation ,Serum Response Factor ,Cellular differentiation ,Histone Deacetylase 1 ,Mice, Inbred Strains ,macromolecular substances ,Regulatory Sequences, Nucleic Acid ,Biology ,Methylation ,Histone Deacetylases ,Mice ,Histone methylation ,Genetics ,Transcriptional regulation ,Animals ,Enhancer of Zeste Homolog 2 Protein ,RNA, Small Interfering ,Muscle, Skeletal ,YY1 Transcription Factor ,MyoD Protein ,Regulation of gene expression ,YY1 ,Myogenesis ,Lysine ,EZH2 ,Polycomb Repressive Complex 2 ,Gene Expression Regulation, Developmental ,Proteins ,Cell Differentiation ,Extremities ,Histone-Lysine N-Methyltransferase ,Methyltransferases ,Research Papers ,Molecular biology ,Chromatin ,Protein Structure, Tertiary ,DNA-Binding Proteins ,embryonic structures ,Erythroid-Specific DNA-Binding Factors ,Transcription Factors ,Developmental Biology - Abstract
The Ezh2 protein endows the Polycomb PRC2 and PRC3 complexes with histone lysine methyltransferase (HKMT) activity that is associated with transcriptional repression. We report that Ezh2 expression was developmentally regulated in the myotome compartment of mouse somites and that its down-regulation coincided with activation of muscle gene expression and differentiation of satellite-cell-derived myoblasts. Increased Ezh2 expression inhibited muscle differentiation, and this property was conferred by its SET domain, required for the HKMT activity. In undifferentiated myoblasts, endogenous Ezh2 was associated with the transcriptional regulator YY1. Both Ezh2 and YY1 were detected, with the deacetylase HDAC1, at genomic regions of silent muscle-specific genes. Their presence correlated with methylation of K27 of histone H3. YY1 was required for Ezh2 binding because RNA interference of YY1 abrogated chromatin recruitment of Ezh2 and prevented H3-K27 methylation. Upon gene activation, Ezh2, HDAC1, and YY1 dissociated from muscle loci, H3-K27 became hypomethylated and MyoD and SRF were recruited to the chromatin. These findings suggest the existence of a two-step activation mechanism whereby removal of H3-K27 methylation, conferred by an active Ezh2-containing protein complex, followed by recruitment of positive transcriptional regulators at discrete genomic loci are required to promote muscle gene expression and cell differentiation.
- Published
- 2004
- Full Text
- View/download PDF
39. Regulation of the p300 HAT domain via a novel activation loop
- Author
-
Vittorio Sartorelli, Jiemin Wong, Natalia Pediconi, Qingyuan Ge, Ling Wang, Marcella Fulco, Robert G. Roeder, Dianzheng Zhang, Paul R. Thompson, Dongxia Wang, Philip A. Cole, Massimo Levrero, Woojin An, and Robert J. Cotter
- Subjects
Enzymologic ,Lysine Acetyltransferases ,Molecular Sequence Data ,Protein domain ,Lysine ,Sequence Homology ,Biology ,Gene Expression Regulation, Enzymologic ,Acetyl Coenzyme A ,Acetyltransferases ,Structural Biology ,Acetylation ,Amino Acid Sequence ,Animals ,Cloning, Molecular ,Conserved Sequence ,DNA Primers ,Enzyme Activation ,Escherichia coli ,Escherichia coli Proteins ,Histone Acetyltransferases ,Kinetics ,Recombinant Proteins ,Sequence Alignment ,Sequence Homology, Amino Acid ,parasitic diseases ,Gene expression ,Molecular Biology ,Kinase ,Autophosphorylation ,Molecular ,Histone acetyltransferase ,Cell biology ,Amino Acid ,Gene Expression Regulation ,Biochemistry ,Activation loop ,biology.protein ,Cloning - Abstract
The transcriptional coactivator p300 is a histone acetyltransferase (HAT) whose function is critical for regulating gene expression in mammalian cells. However, the molecular events that regulate p300 HAT activity are poorly understood. We evaluated autoacetylation of the p300 HAT protein domain to determine its function. Using expressed protein ligation, the p300 HAT protein domain was generated in hypoacetylated form and found to have reduced catalytic activity. This basal catalytic rate was stimulated by autoacetylation of several key lysine sites within an apparent activation loop motif. This post-translational modification and catalytic regulation of p300 HAT activity is conceptually analogous to the activation of most protein kinases by autophosphorylation. We therefore propose that this autoregulatory loop could influence the impact of p300 on a wide variety of signaling and transcriptional events.
- Published
- 2004
- Full Text
- View/download PDF
40. S6K1 ing to Res TOR Adipogenesis with Polycomb
- Author
-
Aster H. Juan and Vittorio Sartorelli
- Subjects
0301 basic medicine ,Genetics ,Adipogenesis ,Kinase ,TOR Serine-Threonine Kinases ,Polycomb-Group Proteins ,P70-S6 Kinase 1 ,Cell Biology ,mTORC1 ,Biology ,Chromatin ,Epigenesis, Genetic ,Serine ,03 medical and health sciences ,030104 developmental biology ,Regulatory sequence ,Animals ,Drosophila Proteins ,Epigenetics ,Molecular Biology - Abstract
Signal-directed chromatin recruitment of mammalian Polycomb complexes is a fundamental component of epigenetic regulation. In this issue, Yi et al. (2016) reveal how mTORC1 activation deploys the ribosomal serine/threonine kinase S6K1 and Polycomb proteins at genomic regulatory regions to repress expression of anti-adipogenic developmental regulators.
- Published
- 2016
- Full Text
- View/download PDF
41. Slug Is a Novel Downstream Target of MyoD
- Author
-
Vittorio Sartorelli, Ethan A. Carver, Eric P. Hoffman, Simona Iezzi, Devin Dressman, Thomas Gridley, and Po Zhao
- Subjects
Zinc finger ,Mesoderm ,animal structures ,biology ,PITX2 ,Slug ,fungi ,Ectoderm ,Cell Biology ,MyoD ,biology.organism_classification ,Biochemistry ,Molecular biology ,Gene expression profiling ,medicine.anatomical_structure ,MyoD Protein ,embryonic structures ,medicine ,Molecular Biology - Abstract
Temporal expression profiling was utilized to define transcriptional regulatory pathways in vivo in a mouse muscle regeneration model. Potential downstream targets of MyoD were identified by temporal expression, promoter data base mining, and gel shift assays; Slug and calpain 6 were identified as novel MyoD targets. Slug, a member of the snail/slug family of zinc finger transcriptional repressors critical for mesoderm/ectoderm development, was further shown to be a downstream target by using promoter/reporter constructs and demonstration of defective muscle regeneration in Slug null mice.
- Published
- 2002
- Full Text
- View/download PDF
42. HERP, a Novel Heterodimer Partner of HES/E(spl) in Notch Signaling
- Author
-
Hung-Yi Wu, Larry Kedes, Tatsuya Iso, Gene Chung, Simona Iezzi, Coralie Poizat, Vittorio Sartorelli, and Yasuo Hamamori
- Subjects
Transcription, Genetic ,Recombinant Fusion Proteins ,Molecular Sequence Data ,Notch signaling pathway ,Repressor ,Biology ,Histone Deacetylases ,Mice ,Chlorocebus aethiops ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Humans ,Nuclear Receptor Co-Repressor 1 ,Amino Acid Sequence ,HEY2 ,Molecular Biology ,Transcription factor ,Psychological repression ,reproductive and urinary physiology ,Homeodomain Proteins ,Transcriptional Regulation ,Binding Sites ,Receptors, Notch ,Basic helix-loop-helix ,Helix-Loop-Helix Motifs ,Membrane Proteins ,Nuclear Proteins ,Proteins ,3T3 Cells ,DNA ,Cell Biology ,Molecular biology ,Repressor Proteins ,Solutions ,DNA binding site ,Sin3 Histone Deacetylase and Corepressor Complex ,Gene Expression Regulation ,COS Cells ,Transcription Factor HES-1 ,biological phenomena, cell phenomena, and immunity ,Co-Repressor Proteins ,Dimerization ,Corepressor ,HeLa Cells ,Mi-2 Nucleosome Remodeling and Deacetylase Complex ,Signal Transduction ,Transcription Factors - Abstract
HERP1 and -2 are members of a new basic helix-loop-helix (bHLH) protein family closely related to HES/E(spl), the only previously known Notch effector. Like that of HES, HERP mRNA expression is directly up-regulated by Notch ligand binding without de novo protein synthesis. HES and HERP are individually expressed in certain cells, but they are also coexpressed within single cells after Notch stimulation. Here, we show that HERP has intrinsic transcriptional repression activity. Transcriptional repression by HES/E(spl) entails the recruitment of the corepressor TLE/Groucho via a conserved WRPW motif, whereas unexpectedly the corresponding—but modified—tetrapeptide motif in HERP confers marginal repression. Rather, HERP uses its bHLH domain to recruit the mSin3 complex containing histone deacetylase HDAC1 and an additional corepressor, N-CoR, to mediate repression. HES and HERP homodimers bind similar DNA sequences, but with distinct sequence preferences, and they repress transcription from specific DNA binding sites. Importantly, HES and HERP associate with each other in solution and form a stable HES-HERP heterodimer upon DNA binding. HES-HERP heterodimers have both a greater DNA binding activity and a stronger repression activity than do the respective homodimers. Thus, Notch signaling relies on cooperation between HES and HERP, two transcriptional repressors with distinctive repression mechanisms which, either as homo- or as heterodimers, regulate target gene expression.
- Published
- 2001
- Full Text
- View/download PDF
43. Regulation of muscle regulatory factors by DNA-binding, interacting proteins, and post-transcriptional modifications
- Author
-
Vittorio Sartorelli and Pier Lorenzo Puri
- Subjects
Physiology ,Myocyte proliferation ,Clinical Biochemistry ,Cell ,Skeletal muscle ,Cell Biology ,Biology ,Cell biology ,Muscle regeneration ,chemistry.chemical_compound ,medicine.anatomical_structure ,Molecular level ,Biochemistry ,chemistry ,Transcription (biology) ,Myogenic regulatory factors ,medicine ,DNA - Abstract
Skeletal muscle differentiation is influenced by multiple pathways, which regulate the activity of myogenic regulatory factors (MRFs)—the myogenic basic helix-loop-helix proteins and the MEF2-family members—in positive or negative ways. Here we will review and discuss the network of signals that regulate MRF function during myocyte proliferation, differentiation, and post-mitotic growth. Elucidating the mechanisms governing muscle-specific transcription will provide important insight in better understanding the embryonic development of muscle at the molecular level and will have important implications in setting out strategies aimed at muscle regeneration. Since the activity of MRFs are compromised in tumors of myogenic derivation—the rhabdomyosarcomas—the studies summarized in this review can provide a useful tool to uncover the molecular basis underlying the formation of these tumors. J. Cell. Physiol. 185:155–173, 2000. © 2000 Wiley-Liss, Inc.
- Published
- 2000
- Full Text
- View/download PDF
44. Regulation of Histone Acetyltransferases p300 and PCAF by the bHLH Protein Twist and Adenoviral Oncoprotein E1A
- Author
-
Yoshihiro Nakatani, Jean Y. J. Wang, Pier Lorenzo Puri, Vittorio Sartorelli, Larry Kedes, Yasuo Hamamori, Hung-Yi Wu, and Vasily Ogryzko
- Subjects
Saccharomyces cerevisiae Proteins ,Transcription, Genetic ,animal diseases ,Biology ,environment and public health ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Acetyltransferases ,Transcription (biology) ,parasitic diseases ,Transcriptional regulation ,Animals ,Cells, Cultured ,Histone Acetyltransferases ,Biochemistry, Genetics and Molecular Biology(all) ,Twist-Related Protein 1 ,Nuclear Proteins ,Oncogene Proteins, Viral ,Molecular biology ,Peptide Fragments ,Cell biology ,Chromatin ,Enzyme Activation ,N-terminus ,enzymes and coenzymes (carbohydrates) ,PCAF ,Acetyltransferase ,COS Cells ,embryonic structures ,Trans-Activators ,Adenovirus E1A Proteins ,E1A-Associated p300 Protein ,Transcription Factors - Abstract
Histone acetyltransferases (HAT) play a critical role in transcriptional control by relieving repressive effects of chromatin, and yet how HATs themselves are regulated remains largely unknown. Here, it is shown that Twist directly binds two independent HAT domains of acetyltransferases, p300 and p300/CBP–associated factor (PCAF), and directly regulates their HAT activities. The N terminus of Twist is a primary domain interacting with both acetyltransferases, and the same domain is required for inhibition of p300-dependent transcription by Twist. Adenovirus E1A protein mimics the effects of Twist by inhibiting the HAT activities of p300 and PCAF. These findings establish a cogent argument for considering the HAT domains as a direct target for acetyltransferase regulation by both a cellular transcription factor and a viral oncoprotein.
- Published
- 1999
- Full Text
- View/download PDF
45. The orphan nuclear receptor, COUP-TF II, inhibits myogenesis by post-transcriptional regulation of MyoD function: COUP-TF II directly interacts with p300 and MyoD
- Author
-
Vittorio Sartorelli, Yasou Hamamori, Peter Bailey, and George E.O. Muscat
- Subjects
Male ,Transcriptional Activation ,Receptors, Steroid ,Biology ,MyoD ,Binding, Competitive ,Mice ,MyoD Protein ,Genes, Reporter ,Coactivator ,Tumor Cells, Cultured ,Genetics ,Animals ,Humans ,RNA Processing, Post-Transcriptional ,Muscle, Skeletal ,Transcription factor ,Cells, Cultured ,Mice, Inbred C3H ,PITX2 ,Myogenesis ,Gene Expression Regulation, Developmental ,Nuclear Proteins ,Cell Differentiation ,3T3 Cells ,Molecular biology ,Peptide Fragments ,Neuron-derived orphan receptor 1 ,DNA-Binding Proteins ,Repressor Proteins ,COUP Transcription Factors ,Nuclear receptor ,Trans-Activators ,E1A-Associated p300 Protein ,Research Article ,Transcription Factors - Abstract
COUP-TF II is an orphan nuclear receptor that has no known ligand in the 'classical sense'. COUP-TF interacts with the corepressors N-CoR, SMRT and RIP13, and silences transcription by active repression and trans-repression. Forced expression of the orphan nuclear receptor COUP-TF II in mouse C2 myogenic cells has been demonstrated to inhibit morphological differentiation, and to repress the expression of: (i) the myoD gene family which encodes myogenic basic helix-loop-helix (bHLH) proteins; and (ii) the cell cycle regulator, p21(Waf-1/Cip-1). In the present study, we show that COUP-TF II efficiently inhibits the myoD -mediated myogenic conversion of pluripotential C3H10T1/2 cells by post-transcriptional mechanisms. Furthermore, repression of MyoD-dependent transcription by COUP-TF II occurs in the absence of the nuclear receptor cognate binding motif. The inhibition of MyoD-mediated trans-activation involves the direct binding of the DNA binding domain/C-region and hinge/D-regions [i.e. amino acid (aa) residues 78-213] of COUP-TF II to the N-terminal activation domain of MyoD. Over-expression of the cofactor p300, which functions as a coactivator of myoD-mediated transcription, alleviated repression by COUP-TF II. Further binding analysis demonstrated that COUP-TF II interacted with the N-terminal 149 aa residues of p300 which encoded the receptor interaction domain of the coactivator. Finally we observed that COUP-TF II, MyoD and p300 interact in a competitive manner, and that increasing amounts of COUP-TF II have the ability to reduce the interaction between myoD and p300 invitro. The experiments presented herein suggest thatCOUP-TF II post-transcriptionally regulates myoD activity/function, and that crosstalk between orphan nuclear receptors and the myogenic bHLH proteins has functional consequences for differentiation.
- Published
- 1998
- Full Text
- View/download PDF
46. Differential Roles of p300 and PCAF Acetyltransferases in Muscle Differentiation
- Author
-
Bruce H. Howard, Pier Lorenzo Puri, Vittorio Sartorelli, Xiang Jiao Yang, Vasily Ogryzko, Adolf Graessmann, Larry Kedes, Massimo Levrero, Yasuo Hamamori, Jean Y. J. Wang, and Yoshihiro Nakatani
- Subjects
Transcriptional Activation ,Saccharomyces cerevisiae Proteins ,Multiprotein complex ,Muscle Fibers, Skeletal ,Cell Cycle Proteins ,MyoD ,Gene Expression Regulation, Enzymologic ,Mice ,Acetyltransferases ,Multienzyme Complexes ,Transcription (biology) ,Animals ,Histone acetyltransferase activity ,p300-CBP Transcription Factors ,Antigens, Viral, Tumor ,Muscle, Skeletal ,Molecular Biology ,Microinjection ,Cells, Cultured ,Histone Acetyltransferases ,MyoD Protein ,biology ,Gene Expression Regulation, Developmental ,Nuclear Proteins ,Cell Differentiation ,Cell Biology ,Histone acetyltransferase ,Acetyl-CoA C-Acyltransferase ,CREB-Binding Protein ,Molecular biology ,PCAF ,Trans-Activators ,biology.protein ,RNA Polymerase II ,E1A-Associated p300 Protein ,Transcription Factors - Abstract
PCAF is a histone acetyltransferase that associates with p300/CBP and competes with E1A for access to them. While exogenous expression of PCAF potentiates both MyoD-directed transcription and myogenic differentiation, PCAF inactivation by anti-PCAF antibody microinjection prevents differentiation. MyoD interacts directly with both p300/CBP and PCAF, forming a multimeric protein complex on the promoter elements. Viral transforming factors that interfere with muscle differentiation disrupt this complex without affecting the MyoD–DNA interaction, indicating functional significance of the complex formation. Exogenous expression of PCAF or p300 promotes p21 expression and terminal cell-cycle arrest. Both of these activities are dependent on the histone acetyltransferase activity of PCAF, but not on that of p300. These results indicate that recruitment of histone acetyltransferase activity of PCAF by MyoD, through p300/CBP, is crucial for activation of the myogenic program.
- Published
- 1997
- Full Text
- View/download PDF
47. Gene transfer and cell transplant: an experimental approach to repair a 'broken heart'
- Author
-
Vittorio Sartorelli, Howard Prentice, Laurence Kedes, Manuel J. Quiñones, Michael Patterson, Robert A. Kloner, and Jonathan Leor
- Subjects
medicine.medical_specialty ,Heart disease ,Cell Transplantation ,Physiology ,business.industry ,Myocardium ,Regeneration (biology) ,Genetic transfer ,Gene Transfer Techniques ,Myocardial Infarction ,Infarction ,Genetic Therapy ,Broken heart ,medicine.disease ,Physiology (medical) ,Heart failure ,Internal medicine ,medicine ,Cardiology ,Humans ,Myocardial infarction ,Cardiology and Cardiovascular Medicine ,business ,Ventricular remodeling ,MyoD Protein - Abstract
Time for primary review 22 days. Despite significant progress in prevention and therapy of ischemic heart disease, treating patients with heart failure after myocardial infarction remains a major therapeutic challenge. Adult cardiomyocytes cannot regenerate after injury. Therefore, cardiomyocyte loss due to myocardial infarction is irreversible. Currently, congestive heart failure is the only major cardiovascular disorder that is increasing in incidence and mortality [1]. Thus, there is still a need to develop alternative therapeutic strategies to prevent, arrest or reverse congestive heart failure after myocardial infarction. Recent insights into the pathogenesis of myocardial disease and advances in molecular biology have opened up a new era of molecular and cellular therapies that target genes, molecules and peptides. Gene transfer as a therapeutic approach for the treatment of myocardial infarction and congestive heart failure has been suggested as a new treatment strategy for these serious disorders [2, 3]. The introduction, into injured myocardium, of recombinant transgenes that encode growth factors, could stimulate new vessel formation with increased collateral blood flow, accelerate healing and enhance myocardial performance. Today it is even possible to consider genetic manipulation leading to regeneration of myocytes within the infarcted myocardium. An alternative experimental strategy to increase viability and augment ventricular function after myocardial infarction is cell transplantation [4]. Engrafted fetal cells might increase the number of functional myocytes in the infarcted myocardium, could serve as a potential source of growth factors and could be programmed for myocyte-based gene transfer. The purpose of our review is to summarize recent advances in the attempts to develop molecular and cellular strategies for repairing a ‘broken heart’. The biological rationale for these new therapies and the potential limitations of gene therapy and cell transplant in treating myocardial infarction are discussed. Left ventricular remodeling following myocardial infarction refers to the process … * Corresponding author. The Heart Institute, Good Samaritan Hospital, 1225 Wilshire Blvd., Los Angeles, CA 90017, USA. Tel.: +1 (213) 977 4050; fax: +1 (213) 977 4107.
- Published
- 1997
- Full Text
- View/download PDF
48. Molecular Mechanisms of Myogenic Coactivation by p300: Direct Interaction with the Activation Domain of MyoD and with the MADS Box of MEF2C
- Author
-
Vittorio Sartorelli, Jing Huang, Yasuo Hamamori, and Larry Kedes
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,animal structures ,Muscle Fibers, Skeletal ,Cell Cycle Proteins ,MADS Domain Proteins ,P300-CBP Transcription Factors ,MyoD ,Cell Line ,Mice ,MyoD Protein ,Acetyltransferases ,Cyclins ,Animals ,Humans ,p300-CBP Transcription Factors ,RNA, Messenger ,CREB-binding protein ,Creatine Kinase ,Molecular Biology ,Myogenin ,Histone Acetyltransferases ,TATA-Binding Protein Associated Factors ,biology ,PITX2 ,MEF2 Transcription Factors ,Myogenesis ,Muscles ,Helix-Loop-Helix Motifs ,Nuclear Proteins ,Cell Differentiation ,Herpes Simplex Virus Protein Vmw65 ,3T3 Cells ,Cell Biology ,musculoskeletal system ,CREB-Binding Protein ,Molecular biology ,Actins ,Cell biology ,DNA-Binding Proteins ,Myogenic Regulatory Factors ,Mutation ,Myogenic regulatory factors ,Trans-Activators ,biology.protein ,Transcription Factor TFIID ,Research Article ,Transcription Factors - Abstract
By searching for molecules that assist MyoD in converting fibroblasts to muscle cells, we have found that p300 and CBP, two related molecules that act as transcriptional adapters, coactivate the myogenic basic-helix-loop-helix (bHLH) proteins. Coactivation by p300 involves novel physical interactions between p300 and the amino-terminal activation domain of MyoD. In particular, disruption of the FYD domain, a group of three amino acids conserved in the activation domains of other myogenic bHLH proteins, drastically diminishes the transactivation potential of MyoD and abolishes both p300-mediated coactivation and the physical interaction between MyoD and p300. Two domains of p300, at its amino and carboxy terminals, independently function to both mediate coactivation and physically interact with MyoD. A truncated segment of p300, unable to bind MyoD, acts as a dominant negative mutation and abrogates both myogenic conversion and transactivation by MyoD, suggesting that endogenous p300 is a required coactivator for MyoD function. The p300 dominant negative peptide forms multimers with intact p300. p300 and CBP serve as coactivators of another class of transcriptional activators critical for myogenesis, myocyte enhancer factor 2 (MEF2). In fact, transactivation mediated by the MEF2C protein is potentiated by the two coactivators, and this phenomenon is associated with the ability of p300 to interact with the MADS domain of MEF2C. Our results suggest that p300 and CBP may positively influence myogenesis by reinforcing the transcriptional autoregulatory loop established between the myogenic bHLH and the MEF2 factors.
- Published
- 1997
- Full Text
- View/download PDF
49. An evolutionarily biased distribution of miRNA sites toward regulatory genes with high promoter-driven intrinsic transcriptional noise
- Author
-
Hossein Zare, Vittorio Sartorelli, and Arkady B. Khodursky
- Subjects
Untranslated region ,Transcription, Genetic ,Evolution ,Biology ,Genome ,Evolution, Molecular ,Mice ,Gene expression ,Genes, Regulator ,medicine ,Transcriptional noise ,Animals ,Humans ,RNA, Messenger ,Promoter Regions, Genetic ,Gene ,Transcription factor ,3' Untranslated Regions ,Ecology, Evolution, Behavior and Systematics ,Regulator gene ,Genetics ,Regulation of gene expression ,Genes, Essential ,medicine.disease ,Biological Evolution ,microRNAs ,Gene Expression Regulation ,Nucleosome occupancy ,Transcription Factors ,Research Article - Abstract
Background miRNAs are a major class of regulators of gene expression in metazoans. By targeting cognate mRNAs, miRNAs are involved in regulating most, if not all, biological processes in different cell and tissue types. To better understand how this regulatory potential is allocated among different target gene sets, we carried out a detailed and systematic analysis of miRNA target sites distribution in the mouse genome. Results We used predicted conserved and non-conserved sites for 779 miRNAs in 3′ UTR of 18440 genes downloaded from TargetScan website. Our analysis reveals that 3′ UTRs of genes encoding regulatory proteins harbor significantly greater number of miRNA sites than those of non-regulatory, housekeeping and structural, genes. Analysis of miRNA sites for orthologous 3′UTR’s in 10 other species indicates that the regulatory genes were maintaining or accruing miRNA sites while non-regulatory genes gradually shed them in the course of evolution. Furthermore, we observed that 3′ UTR of genes with higher gene expression variability driven by their promoter sequence content are targeted by many more distinct miRNAs compared to genes with low transcriptional noise. Conclusions Based on our results we envision a model, which we dubbed “selective inclusion”, whereby non-regulatory genes with low transcription noise and stable expression profile lost their sites, while regulatory genes which endure higher transcription noise retained and gained new sites. This adaptation is consistent with the requirements that regulatory genes need to be tightly controlled in order to have precise and optimum protein level to properly function.
- Published
- 2013
50. Author response: H3K4 mono- and di-methyltransferase MLL4 is required for enhancer activation during cell differentiation
- Author
-
Vittorio Sartorelli, Weiqun Peng, Lifeng Wang, Ji-Eun Lee, Kai Ge, Anne Baldridge, Lenan Zhuang, Shiliyang Xu, Chaochen Wang, Xuesong Feng, and Young Wook Cho
- Subjects
Methyltransferase ,Cellular differentiation ,Biology ,Enhancer ,Cell biology - Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.