Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Nucleic Acids Res ; 47(3): 1278-1293, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30544196

ABSTRACT

SMYD3 is a methylase previously linked to cancer cell invasion and migration. Here we show that SMYD3 favors TGFß-induced epithelial-mesenchymal transition (EMT) in mammary epithelial cells, promoting mesenchymal and EMT transcription factors expression. SMYD3 directly interacts with SMAD3 but it is unnecessary for SMAD2/3 phosphorylation and nuclear translocation. Conversely, SMYD3 is indispensable for SMAD3 direct association to EMT genes regulatory regions. Accordingly, SMYD3 knockdown or its pharmacological blockade with the BCI121 inhibitor dramatically reduce TGFß-induced SMAD3 association to the chromatin. Remarkably, BCI121 treatment attenuates mesenchymal genes transcription in the mesenchymal-like MDA-MB-231 cell line and reduces their invasive ability in vivo, in a zebrafish xenograft model. In addition, clinical datasets analysis revealed that higher SMYD3 levels are linked to a less favorable prognosis in claudin-low breast cancers and to a reduced metastasis free survival in breast cancer patients. Overall, our data point at SMYD3 as a pivotal SMAD3 cofactor that promotes TGFß-dependent mesenchymal gene expression and cell migration in breast cancer, and support SMYD3 as a promising pharmacological target for anti-cancer therapy.


Subject(s)
Breast Neoplasms/genetics , Histone-Lysine N-Methyltransferase/genetics , Smad3 Protein/genetics , Transforming Growth Factor beta/genetics , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Chromatin/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Knockdown Techniques , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Phosphorylation , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , Zebrafish
2.
Genes Dev ; 27(11): 1299-312, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23752591

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.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Myostatin/genetics , Positive Transcriptional Elongation Factor B/metabolism , Proto-Oncogene Proteins c-met/genetics , Animals , Cell Line , Cyclin-Dependent Kinase 9/metabolism , Dexamethasone/pharmacology , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Mice , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscle Proteins/genetics , Muscle, Skeletal/drug effects , Muscular Atrophy/chemically induced , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Phosphorylation , Phosphoserine/metabolism , Protein Binding , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , SKP Cullin F-Box Protein Ligases/genetics , Transcription Factors/chemistry , Transcription Factors/metabolism , Transcription, Genetic
3.
J Cell Physiol ; 230(10): 2447-2460, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25728514

ABSTRACT

SMYD3 is a histone lysine methyltransferase that plays an important role in transcriptional activation as a member of an RNA polymerase complex, and its oncogenic role has been described in different cancer types. We studied the expression and activity of SMYD3 in a preclinical model of colorectal cancer (CRC) and found that it is strongly upregulated throughout tumorigenesis both at the mRNA and protein level. Our results also showed that RNAi-mediated SMYD3 ablation impairs CRC cell proliferation indicating that SMYD3 is required for proper cancer cell growth. These data, together with the importance of lysine methyltransferases as a target for drug discovery, prompted us to carry out a virtual screening to identify new SMYD3 inhibitors by testing several candidate small molecules. Here we report that one of these compounds (BCI-121) induces a significant reduction in SMYD3 activity both in vitro and in CRC cells, as suggested by the analysis of global H3K4me2/3 and H4K5me levels. Of note, the extent of cell growth inhibition by BCI-121 was similar to that observed upon SMYD3 genetic ablation. Most of the results described above were obtained in CRC; however, when we extended our observations to tumor cell lines of different origin, we found that SMYD3 inhibitors are also effective in other cancer types, such as lung, pancreatic, prostate, and ovarian. These results represent the proof of principle that SMYD3 is a druggable target and suggest that new compounds capable of inhibiting its activity may prove useful as novel therapeutic agents in cancer treatment.


Subject(s)
Cell Proliferation/drug effects , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/metabolism , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/pathology , Mice , RNA Interference/drug effects , Transcriptional Activation/drug effects , Up-Regulation
4.
Comp Funct Genomics ; 2012: 979168, 2012.
Article in English | MEDLINE | ID: mdl-22701348

ABSTRACT

Recent advances in high-throughput technologies have transformed methodologies employed to study cell-specific epigenomes and the approaches to investigate complex cellular phenotypes. Application of next-generation sequencing technology in the skeletal muscle differentiation field is rapidly extending our knowledge on how chromatin modifications, transcription factors and chromatin regulators orchestrate gene expression pathways guiding myogenesis. Here, we review recent biological insights gained by the application of next-generation sequencing techniques to decode the epigenetic profile and gene regulatory networks underlying skeletal muscle differentiation.

5.
Cells ; 10(5)2021 05 18.
Article in English | MEDLINE | ID: mdl-34069776

ABSTRACT

SMYD3 (SET and MYND domain containing protein 3) is a methylase over-expressed in cancer cells and involved in oncogenesis. While several studies uncovered key functions for SMYD3 in cancer models, the SMYD3 role in physiological conditions has not been fully elucidated yet. Here, we dissect the role of SMYD3 at early stages of development, employing mouse embryonic stem cells (ESCs) and zebrafish as model systems. We report that SMYD3 depletion promotes the induction of the mesodermal pattern during in vitro differentiation of ESCs and is linked to an upregulation of cardiovascular lineage markers at later stages. In vivo, smyd3 knockdown in zebrafish favors the upregulation of mesendodermal markers during zebrafish gastrulation. Overall, our study reveals that SMYD3 modulates levels of mesendodermal markers, both in development and in embryonic stem cell differentiation.


Subject(s)
Cell Differentiation , Histone-Lysine N-Methyltransferase/metabolism , Mouse Embryonic Stem Cells/enzymology , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cell Line , Cell Lineage , Embryonic Development , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/genetics , Mice , Time Factors , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
6.
Nat Commun ; 11(1): 6108, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33257646

ABSTRACT

Duchenne muscular dystrophy (DMD) affects 1 in 3500 live male births. To date, there is no effective cure for DMD, and the identification of novel molecular targets involved in disease progression is important to design more effective treatments and therapies to alleviate DMD symptoms. Here, we show that protein levels of the Bromodomain and extra-terminal domain (BET) protein BRD4 are significantly increased in the muscle of the mouse model of DMD, the mdx mouse, and that pharmacological inhibition of the BET proteins has a beneficial outcome, tempering oxidative stress and muscle damage. Alterations in reactive oxygen species (ROS) metabolism are an early event in DMD onset and they are tightly linked to inflammation, fibrosis, and necrosis in skeletal muscle. By restoring ROS metabolism, BET inhibition ameliorates these hallmarks of the dystrophic muscle, translating to a beneficial effect on muscle function. BRD4 direct association to chromatin regulatory regions of the NADPH oxidase subunits increases in the mdx muscle and JQ1 administration reduces BRD4 and BRD2 recruitment at these regions. JQ1 treatment reduces NADPH subunit transcript levels in mdx muscles, isolated myofibers and DMD immortalized myoblasts. Our data highlight novel functions of the BET proteins in dystrophic skeletal muscle and suggest that BET inhibitors may ameliorate the pathophysiology of DMD.


Subject(s)
Muscular Dystrophy, Duchenne/metabolism , Nuclear Proteins/metabolism , Oxidative Stress/drug effects , Transcription Factors/metabolism , Animals , Azepines/pharmacology , Disease Models, Animal , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/pathology , NADP , NADPH Oxidases/metabolism , Neuromuscular Diseases/metabolism , Nuclear Proteins/drug effects , Reactive Oxygen Species/metabolism , Transcription Factors/drug effects , Triazoles/pharmacology
7.
Oncotarget ; 8(13): 21778-21793, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28423519

ABSTRACT

Low circulating levels of vitamin D were associated with decreased muscle strength and physical performance. Along this line, the present study was aimed to investigate: i) the therapeutic potential of vitamin D in cancer-induced muscle wasting; ii) the mechanisms by which vitamin D affects muscle phenotype in tumor-bearing animals.Rats bearing the AH130 hepatoma showed decreased circulating vitamin D compared to control rats, while muscle vitamin D receptor (VDR) mRNA was up-regulated. Both circulating vitamin D and muscle VDR expression increased after vitamin D administration, without exerting appreciable effects on body weight and muscle mass.The effects of vitamin D on muscle cells were studied in C2C12 myocytes. Vitamin D-treated myoblasts did not differentiate properly, fusing only partially and forming multinucleated structures with aberrant shape and low myosin heavy chain content. Vitamin D treatment resulted in VDR overexpression and myogenin down-regulation. Silencing VDR expression in C2C12 cultures abrogated the inhibition of differentiation exerted by vitamin D treatment.These data suggest that VDR overexpression in tumor-bearing animals contributes to muscle wasting by impairing muscle regenerative program. In this regard, attention should be paid when considering vitamin D supplementation to patients affected by chronic pathologies where muscle regeneration may be involved.


Subject(s)
Cachexia/metabolism , Muscle, Skeletal/metabolism , Receptors, Calcitriol/metabolism , Vitamin D/metabolism , Animals , Blotting, Western , Cachexia/etiology , Carcinoma, Hepatocellular/complications , Cell Line , Chromatin Immunoprecipitation , Disease Models, Animal , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Liver Neoplasms/complications , Mice, Inbred BALB C , Mice, Inbred C57BL , Muscle Development/drug effects , Muscle, Skeletal/drug effects , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Regeneration/drug effects , Vitamin D/pharmacology
8.
Nat Commun ; 8(1): 1707, 2017 11 22.
Article in English | MEDLINE | ID: mdl-29167426

ABSTRACT

Cancer cachexia is a devastating metabolic syndrome characterized by systemic inflammation and massive muscle and adipose tissue wasting. Although it is responsible for approximately one-third of cancer deaths, no effective therapies are available and the underlying mechanisms have not been fully elucidated. We previously identified the bromodomain and extra-terminal domain (BET) protein BRD4 as an epigenetic regulator of muscle mass. Here we show that the pan-BET inhibitor (+)-JQ1 protects tumor-bearing mice from body weight loss and muscle and adipose tissue wasting. Remarkably, in C26-tumor-bearing mice (+)-JQ1 administration dramatically prolongs survival, without directly affecting tumor growth. By ChIP-seq and ChIP analyses, we unveil that BET proteins directly promote the muscle atrophy program during cachexia. In addition, BET proteins are required to coordinate an IL6-dependent AMPK nuclear signaling pathway converging on FoxO3 transcription factor. Overall, these findings indicate that BET proteins may represent a promising therapeutic target in the management of cancer cachexia.


Subject(s)
Cachexia/prevention & control , Neoplasms, Experimental/therapy , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Azepines/pharmacology , Cachexia/genetics , Cachexia/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Epigenesis, Genetic , Forkhead Box Protein O3/metabolism , Gene Expression Regulation , Humans , Interleukin-6/metabolism , Male , Metabolic Networks and Pathways/drug effects , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/prevention & control , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Triazoles/pharmacology
9.
Mol Pharm ; 6(3): 696-705, 2009.
Article in English | MEDLINE | ID: mdl-19267451

ABSTRACT

We have developed lipid-polycation-DNA (LPD) nanoparticles containing DOTAP and targeted with polyethylene glycol (PEG) tethered with anisamide (AA) to specifically deliver siRNA to H460 human lung carcinoma cells which express the sigma receptor. A novel non-glycerol based cationic lipid which contains both a guanidinium and a lysine residue as the cationic headgroup, i.e. DSGLA, downregulated pERK more efficiently in H460 cells than DOTAP. As demonstrated by using fluorescently labeled siRNA, LPD-PEG-AA prepared with DSGLA efficiently delivered siRNA to the cytoplasm of the H460 cells. Although the siRNA delivered by LPD-PEG-AA containing either DOTAP or DSGLA could effectively silence EGFR expression, a synergistic cell killing effect in promoting cellular apoptosis was only observed with DSGLA. The fluorescently labeled siRNA was efficiently delivered into the cytoplasm of H460 xenograft tumor by the LPD-PEG-AA containing either DOTAP or DSGLA 4 h after intravenous injection. Three daily injections (0.6 mg/kg) of siRNA formulated in the LPD-PEG-AA containing either DOTAP or DSGLA could effectively silence the epidermal growth factor receptor (EGFR) in the tumor, but the formulation containing DSGLA could induce more cellular apoptosis. A significant improvement in tumor growth inhibition was observed after dosing with LPD-PEG-AA containing DSGLA. Thus, DSGLA served as both a formulation component as well as a therapeutic agent which synergistically enhanced the activity of siRNA.


Subject(s)
Lipids/chemistry , RNA, Small Interfering/therapeutic use , Small Cell Lung Carcinoma/drug therapy , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , ErbB Receptors/genetics , Female , Gene Silencing , Guanidine/chemistry , Humans , Lysine/chemistry , Mice , Mice, Nude , Microscopy, Fluorescence , Nanoparticles/chemistry , Polyethylene Glycols , RNA, Small Interfering/chemical synthesis , RNA, Small Interfering/pharmacokinetics , RNA, Small Interfering/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL