56 results on '"Caretti, G"'
Search Results
2. 17 The enhancer of the human uPA gene is accessible to trancription factors in both uninduced and TPA-induced HepG2 cells
- Author
-
Ibañez-Tallon, I., Caretti, G., Bonte, E., Blasi, F., and Crippa, M.P.
- Published
- 1997
- Full Text
- View/download PDF
3. TECHNICAL AND PHYSICAL-DOSIMETRIC COMPARISONS BETWEEN A TELECOBALT THERAPY UNIT AND A VAN DE GRAAFF ELECTROSTATIC GENERATOR (2 Mev)
- Author
-
Caretti, G
- Published
- 1961
4. PROBLEMS OF FETAL DOSIMETRY DURING OBSTETRIC X-RAY DIAGNOSIS
- Author
-
Caretti, G
- Published
- 1964
5. The BET inhibitor JQ1 targets fat metabolism and counteracts obesity.
- Author
-
Fornelli C, Sofia Cento A, Nevi L, Mastrocola R, Ferreira Alves G, Caretti G, Collino M, and Penna F
- Subjects
- Animals, Male, Mice, Adipocytes metabolism, Adipocytes drug effects, Muscle, Skeletal metabolism, Muscle, Skeletal drug effects, Adipose Tissue, White metabolism, Adipose Tissue, White drug effects, Transcription Factors metabolism, Transcription Factors antagonists & inhibitors, Humans, Lipid Metabolism drug effects, Disease Models, Animal, Adiposity drug effects, Adipose Tissue metabolism, Adipose Tissue drug effects, Azepines pharmacology, Triazoles pharmacology, Obesity drug therapy, Obesity metabolism, Mice, Inbred C57BL, Diet, High-Fat adverse effects
- Abstract
Introduction: Obesity, one of the most frequent health problems in the adult population, is a condition characterized by excessive white adipose tissue accumulation and accompanied by the increased risk to develop other disorders such as type II diabetes, cardiovascular disorders, physical disability, frailty and sarcopenia. Total fat mass frequently increases during aging, often coexisting with sarcopenia, thus resulting in an emerging condition defined sarcopenic obesity (SO). Our previous data demonstrated the relevant role of the bromo and extra-terminal domain (BET) proteins inhibitor JQ1 in attenuating inflammation and fibrosis in sarcopenic mice. Moreover, we preliminarily observed that JQ1 administration markedly reduces white adipose tissue mass, suggesting a potential role of BET proteins on visceral fat deposition during aging., Objectives: Starting from those observations, the aim of this study was to investigate the ability of JQ1 to reduce adiposity in a chronic diet-induced obesity (DIO) mouse model mimicking the human metabolic syndrome., Methods: Male C57BL/6J mice were divided in subgroups, either fed a standard diet or a high fat diet for 22 or 12 weeks, treated over the last 14 days with JQ1 or with vehicle., Results: The results showed that JQ1 administration reduces fat mass, preserving skeletal muscle mass and function. A direct JQ1 lipolytic effect was demonstrated on mature adipocyte cultures. JQ1-mediated loss of adipose tissue mass was not associated with systemic inflammation or with lipid accumulation in muscle and liver. JQ1 administration did not impinge on skeletal muscle metabolism and oxidative capability, as shown by the lack of significant impact on mitochondrial mass and biogenesis., Conclusion: In conclusion, the current data highlight a potential benefit of JQ1 administration to counteract obesity, suggesting epigenetic modulation as a prospective target in the treatment of obesity and sarcopenic obesity, despite the underlying multiorgan molecular mechanism is still not completely elucidated., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
6. Targeting Epigenetic Regulators with HDAC and BET Inhibitors to Modulate Muscle Wasting.
- Author
-
Nevi L, Pöllänen N, Penna F, and Caretti G
- Subjects
- Humans, Cachexia metabolism, Epigenesis, Genetic, Muscle, Skeletal metabolism, Muscular Atrophy drug therapy, Muscular Atrophy genetics, Proteins metabolism, Histone Deacetylases metabolism, Antineoplastic Agents pharmacology, Neoplasms metabolism, Sarcopenia metabolism
- Abstract
Epigenetic changes contribute to the profound alteration in the transcriptional program associated with the onset and progression of muscle wasting in several pathological conditions. Although HDACs and their inhibitors have been extensively studied in the field of muscular dystrophies, the potential of epigenetic inhibitors has only been marginally explored in other disorders associated with muscle atrophy, such as in cancer cachexia and sarcopenia. BET inhibitors represent a novel class of recently developed epigenetic drugs that display beneficial effects in a variety of diseases beyond malignancies. Based on the preliminary in vitro and preclinical data, HDACs and BET proteins contribute to the pathogenesis of cancer cachexia and sarcopenia, modulating processes related to skeletal muscle mass maintenance and/or metabolism. Thus, epigenetic drugs targeting HDACs and BET proteins may emerge as promising strategies to reverse the catabolic phenotype associated with cachexia and sarcopenia. Further preclinical studies are warranted to delve deeper into the molecular mechanisms associated with the functions of HDACs and BET proteins in muscle atrophy and to establish whether their epigenetic inhibitors represent a prospective therapeutic avenue to alleviate muscle wasting.
- Published
- 2023
- Full Text
- View/download PDF
7. Discovery of the 4-aminopiperidine-based compound EM127 for the site-specific covalent inhibition of SMYD3.
- Author
-
Parenti MD, Naldi M, Manoni E, Fabini E, Cederfelt D, Talibov VO, Gressani V, Guven U, Grossi V, Fasano C, Sanese P, De Marco K, Shtil AA, Kurkin AV, Altieri A, Danielson UH, Caretti G, Simone C, Varchi G, Bartolini M, and Del Rio A
- Subjects
- Humans, Female, Histones, Cell Line, Tumor, Histone-Lysine N-Methyltransferase metabolism, Breast Neoplasms
- Abstract
Recent findings support the hypothesis that inhibition of SMYD3 methyltransferase may be a therapeutic avenue for some of the deadliest cancer types. Herein, active site-selective covalent SMYD3 inhibitors were designed by introducing an appropriate reactive cysteine trap into reversible first-generation SMYD3 inhibitors. The 4-aminopiperidine derivative EM127 (11C) bearing a 2-chloroethanoyl group as reactive warhead showed selectivity for Cys186, located in the substrate/histone binding pocket. Selectivity towards Cys186 was retained even at high inhibitor/enzyme ratio, as shown by mass spectrometry. The mode of interaction with the SMYD3 substrate/histone binding pocket was revealed by crystallographic studies. In enzymatic assays, 11C showed a stronger SMYD3 inhibitory effect compared to the reference inhibitor EPZ031686. Remarkably, 11C attenuated the proliferation of MDA-MB-231 breast cancer cell line at the same low micromolar range of concentrations that reduced SMYD3 mediated ERK signaling in HCT116 colorectal cancer and MDA-MB-231 breast cancer cells. Furthermore, 11C (5 μM) strongly decreased the steady-state mRNA levels of genes important for tumor biology such as cyclin dependent kinase 2, c-MET, N-cadherin and fibronectin 1, all known to be regulated, at least in part, by SMYD3. Thus, 11C is as a first example of second generation SMYD3 inhibitors; this agent represents a covalent and a site specific SMYD3 binder capable of potent and prolonged attenuation of methyltransferase activity., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2022 Elsevier Masson SAS. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
8. Exercise and Exercise Mimetics for the Treatment of Musculoskeletal Disorders.
- Author
-
Cento AS, Leigheb M, Caretti G, and Penna F
- Subjects
- Exercise physiology, Exercise Therapy, Humans, Sedentary Behavior, Musculoskeletal Diseases therapy, Quality of Life
- Abstract
Purpose of Review: The incidence of musculoskeletal disorders affecting bones, joints, and muscles is dramatically increasing in parallel with the increased longevity of the worldwide population, severely impacting on the individual's quality of life and on the healthcare costs. Inactivity and sedentary lifestyle are nowadays considered the main drivers of age-associated musculoskeletal disorders and exercise may counteract such alterations also in other bone- and muscle-centered disorders. This review aims at clarifying the potential use of exercise training to improve musculoskeletal health., Recent Findings: Both the skeletal muscle and the bone are involved in a complex crosstalk determining, in part through tissue-specific and inflammatory/immune released factors, the occurrence of musculoskeletal disorders. Exercise is able to modulate the levels of those molecules and several associated molecular pathways. Evidence from preclinical and clinical trials supports the adoption of exercise and the future use of exercise mimicking drugs will optimize the care of individuals with musculoskeletal disorders., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
9. Sarcopenia Diagnosis: Reliability of the Ultrasound Assessment of the Tibialis Anterior Muscle as an Alternative Evaluation Tool.
- Author
-
Leigheb M, de Sire A, Colangelo M, Zagaria D, Grassi FA, Rena O, Conte P, Neri P, Carriero A, Sacchetti GM, Penna F, Caretti G, and Ferraro E
- Abstract
Sarcopenia is a skeletal muscle disorder characterized by reduced muscle mass, strength, and performance. Muscle ultrasound can be helpful in assessing muscle mass, quality, and architecture, and thus possibly useful for diagnosing or screening sarcopenia. The objective of this study was to evaluate the reliability of ultrasound assessment of tibialis anterior muscle in sarcopenia diagnosis. We included subjects undergoing total or partial hip replacement, comparing measures with a healthy control group. We measured the following parameters: tibialis anterior muscle thickness, echogenicity, architecture, stiffness, skeletal muscle index (SMI), hand grip strength, and sarcopenia related quality of life evaluated through the SarQoL questionnaire. We included 33 participants with a mean age of 54.97 ± 23.91 years. In the study group we found reduced tibialis anterior muscle thickness compared to the healthy control group (19.49 ± 4.92 vs. 28.94 ± 3.63 mm, p < 0.05) with significant correlation with SarQoL values (r = 0.80, p < 0.05), dynamometer hand strength (r = 0.72, p < 0.05) and SMI (r = 0.76, p < 0.05). Moreover, we found reduced stiffness (32.21 ± 12.31 vs. 27.07 ± 8.04 Kpa, p < 0.05). AUC measures of ROC curves were 0.89 predicting reduced muscle strength, and 0.97 predicting reduced SMI for tibialis anterior muscle thickness, while they were 0.73 and 0.85, respectively, for muscle stiffness. Our findings showed that ultrasound assessment of tibialis anterior muscle might be considered a reliable measurement tool to evaluate sarcopenia.
- Published
- 2021
- Full Text
- View/download PDF
10. The Lysine Methylase SMYD3 Modulates Mesendodermal Commitment during Development.
- Author
-
Fittipaldi R, Floris P, Proserpio V, Cotelli F, Beltrame M, and Caretti G
- Subjects
- 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, Cell Differentiation, Histone-Lysine N-Methyltransferase metabolism, Mouse Embryonic Stem Cells enzymology, Zebrafish metabolism, Zebrafish Proteins metabolism
- 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.
- Published
- 2021
- Full Text
- View/download PDF
11. DCLK1, a Putative Stem Cell Marker in Human Cholangiocarcinoma.
- Author
-
Nevi L, Di Matteo S, Carpino G, Zizzari IG, Samira S, Ambrosino V, Costantini D, Overi D, Giancotti A, Monti M, Bosco D, De Peppo V, Oddi A, De Rose AM, Melandro, Bragazzi MC, Faccioli J, Massironi S, Grazi GL, Panici PB, Berloco PB, Giuliante F, Cardinale V, Invernizzi P, Caretti G, Gaudio E, and Alvaro D
- Subjects
- Bile Duct Neoplasms pathology, Biomarkers, Tumor genetics, Cell Line, Tumor, Cell Proliferation, Cholangiocarcinoma pathology, Doublecortin-Like Kinases, Gene Expression Regulation, Neoplastic, Humans, Intracellular Signaling Peptides and Proteins genetics, Neoplastic Stem Cells pathology, Protein Serine-Threonine Kinases genetics, Receptors, G-Protein-Coupled genetics, Bile Duct Neoplasms genetics, Biomarkers, Tumor biosynthesis, Cholangiocarcinoma genetics, Intracellular Signaling Peptides and Proteins biosynthesis, Protein Serine-Threonine Kinases biosynthesis, Receptors, G-Protein-Coupled biosynthesis
- Abstract
Background and Aims: Cholangiocarcinoma (CCA) is a very aggressive cancer showing the presence of high cancer stem cells (CSCs). Doublecortin-like kinase1 (DCLK1) has been demonstrated as a CSC marker in different gastroenterological solid tumors. Our aim was to evaluate in vitro the expression and the biological function of DCLK1 in intrahepatic CCA (iCCA) and perihilar CCA (pCCA)., Approach and Results: Specimens surgically resected of human CCA were enzymatically digested, submitted to immunosorting for specific CSC markers (LGR5 [leucine-rich repeat-containing G protein-coupled receptor], CD [clusters of differentiation] 90, EpCAM [epithelial cell adhesion molecule], CD133, and CD13), and primary cell cultures were prepared. DCLK1 expression was analyzed in CCA cell cultures by real-time quantitative PCR, western blot, and immunofluorescence. Functional studies have been performed by evaluating the effects of selective DCLK1 inhibitor (LRRK2-IN-1) on cell proliferation (MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium] assay, cell population doubling time), apoptosis, and colony formation capacity. DCLK1 was investigated in situ by immunohistochemistry and real-time quantitative PCR. DCLK1 serum concentration was analyzed by enzyme-linked immunosorbent assay. We describe DCLK1 in CCA with an increased gene and protein DCLK1 expression in pCCA
LGR5+ and in iCCACD133+ cells compared with unsorted cells. LRRK2-IN-1 showed an anti-proliferative effect in a dose-dependent manner. LRRK2-IN-1 markedly impaired cell proliferation, induced apoptosis, and decreased colony formation capacity and colony size in both iCCA and pCCA compared with the untreated cells. In situ analysis confirmed that DCLK1 is present only in tumors, and not in healthy tissue. Interestingly, DCLK1 was detected in the human serum samples of patients with iCCA (high), pCCA (high), HCC (low), and cirrhosis (low), but it was almost undetectable in healthy controls., Conclusions: DCLK1 characterizes a specific CSC subpopulation of iCCACD133+ and pCCALGR5+ , and its inhibition exerts anti-neoplastic effects in primary CCA cell cultures. Human DCLK1 serum might represent a serum biomarker for the early CCA diagnosis., (© 2020 by the American Association for the Study of Liver Diseases.)- Published
- 2021
- Full Text
- View/download PDF
12. BETs inhibition attenuates oxidative stress and preserves muscle integrity in Duchenne muscular dystrophy.
- Author
-
Segatto M, Szokoll R, Fittipaldi R, Bottino C, Nevi L, Mamchaoui K, Filippakopoulos P, and Caretti G
- Subjects
- 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, Muscular Dystrophy, Duchenne metabolism, Nuclear Proteins metabolism, Oxidative Stress drug effects, Transcription Factors metabolism
- 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.
- Published
- 2020
- Full Text
- View/download PDF
13. Targeting SMYD3 to Sensitize Homologous Recombination-Proficient Tumors to PARP-Mediated Synthetic Lethality.
- Author
-
Sanese P, Fasano C, Buscemi G, Bottino C, Corbetta S, Fabini E, Silvestri V, Valentini V, Disciglio V, Forte G, Lepore Signorile M, De Marco K, Bertora S, Grossi V, Guven U, Porta N, Di Maio V, Manoni E, Giannelli G, Bartolini M, Del Rio A, Caretti G, Ottini L, and Simone C
- Abstract
SMYD3 is frequently overexpressed in a wide variety of cancers. Indeed, its inactivation reduces tumor growth in preclinical in vivo animal models. However, extensive characterization in vitro failed to clarify SMYD3 function in cancer cells, although confirming its importance in carcinogenesis. Taking advantage of a SMYD3 mutant variant identified in a high-risk breast cancer family, here we show that SMYD3 phosphorylation by ATM enables the formation of a multiprotein complex including ATM, SMYD3, CHK2, and BRCA2, which is required for the final loading of RAD51 at DNA double-strand break sites and completion of homologous recombination (HR). Remarkably, SMYD3 pharmacological inhibition sensitizes HR-proficient cancer cells to PARP inhibitors, thereby extending the potential of the synthetic lethality approach in human tumors., Competing Interests: The authors declare no competing interests., (© 2020 The Author(s).)
- Published
- 2020
- Full Text
- View/download PDF
14. Inhibition of Bromodomain and Extraterminal Domain (BET) Proteins by JQ1 Unravels a Novel Epigenetic Modulation to Control Lipid Homeostasis.
- Author
-
Tonini C, Colardo M, Colella B, Di Bartolomeo S, Berardinelli F, Caretti G, Pallottini V, and Segatto M
- Subjects
- Cell Proliferation drug effects, Cholesterol metabolism, Hep G2 Cells, Humans, Hydroxymethylglutaryl CoA Reductases metabolism, Membrane Proteins metabolism, Phosphorylation, Proteins antagonists & inhibitors, Receptors, LDL metabolism, Sterol Regulatory Element Binding Proteins metabolism, Azepines pharmacology, Epigenesis, Genetic drug effects, Lipid Metabolism drug effects, Proteins metabolism, Triazoles pharmacology
- Abstract
The homeostatic control of lipid metabolism is essential for many fundamental physiological processes. A deep understanding of its regulatory mechanisms is pivotal to unravel prospective physiopathological factors and to identify novel molecular targets that could be employed to design promising therapies in the management of lipid disorders. Here, we investigated the role of bromodomain and extraterminal domain (BET) proteins in the regulation of lipid metabolism. To reach this aim, we used a loss-of-function approach by treating HepG2 cells with JQ1, a powerful and selective BET inhibitor. The main results demonstrated that BET inhibition by JQ1 efficiently decreases intracellular lipid content, determining a significant modulation of proteins involved in lipid biosynthesis, uptake and intracellular trafficking. Importantly, the capability of BET inhibition to slow down cell proliferation is dependent on the modulation of cholesterol metabolism. Taken together, these data highlight a novel epigenetic mechanism involved in the regulation of lipid homeostasis., Competing Interests: The authors declare no conflict of interest.
- Published
- 2020
- Full Text
- View/download PDF
15. SMYD3: An Oncogenic Driver Targeting Epigenetic Regulation and Signaling Pathways.
- Author
-
Bottino C, Peserico A, Simone C, and Caretti G
- Abstract
SMYD3 is a member of the SMYD lysine methylase family and plays an important role in the methylation of various histone and non-histone targets. Aberrant SMYD3 expression contributes to carcinogenesis and SMYD3 upregulation was proposed as a prognostic marker in various solid cancers. Here we summarize SMYD3-mediated regulatory mechanisms, which are implicated in the pathophysiology of cancer, as drivers of distinct oncogenic pathways. We describe SMYD3-dependent mechanisms affecting cancer progression, highlighting SMYD3 interplay with proteins and RNAs involved in the regulation of cancer cell proliferation, migration and invasion. We also address the effectiveness and mechanisms of action for the currently available SMYD3 inhibitors. The findings analyzed herein demonstrate that a complex network of SMYD3-mediated cytoplasmic and nuclear interactions promote oncogenesis across different cancer types. These evidences depict SMYD3 as a modulator of the transcriptional response and of key signaling pathways, orchestrating multiple oncogenic inputs and ultimately, promoting transcriptional reprogramming and tumor transformation. Further insights into the oncogenic role of SMYD3 and its targeting of different synergistic oncogenic signals may be beneficial for effective cancer treatment.
- Published
- 2020
- Full Text
- View/download PDF
16. Interplay between Metabolites and the Epigenome in Regulating Embryonic and Adult Stem Cell Potency and Maintenance.
- Author
-
Harvey A, Caretti G, Moresi V, Renzini A, and Adamo S
- Subjects
- Animals, Biomarkers, Disease Models, Animal, Gene Expression Regulation, Developmental, Humans, Phenotype, Stem Cell Transplantation, Adult Stem Cells cytology, Adult Stem Cells metabolism, Cell Differentiation genetics, Cell Self Renewal genetics, Embryonic Stem Cells cytology, Embryonic Stem Cells metabolism, Energy Metabolism, Epigenesis, Genetic
- Abstract
The environment surrounding stem cells has the ability to elicit profound, heritable epigenetic changes orchestrated by multiple epigenetic mechanisms, which can be modulated by the level of specific metabolites. In this review, we highlight the significance of metabolism in regulating stem cell homeostasis, cell state, and differentiation capacity, using metabolic regulation of embryonic and adult muscle stem cells as examples, and cast light on the interaction between cellular metabolism and epigenetics. These new regulatory networks, based on the dynamic interplay between metabolism and epigenetics in stem cell biology, are important, not only for understanding tissue homeostasis, but to determine in vitro culture conditions which accurately support normal cell physiology., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
17. Metabolic Control of Stemness and Differentiation.
- Author
-
Moresi V, Sotiropoulos A, Caretti G, and Harvey A
- Published
- 2019
- Full Text
- View/download PDF
18. SMYD3 promotes the epithelial-mesenchymal transition in breast cancer.
- Author
-
Fenizia C, Bottino C, Corbetta S, Fittipaldi R, Floris P, Gaudenzi G, Carra S, Cotelli F, Vitale G, and Caretti G
- Subjects
- 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, Breast Neoplasms genetics, Histone-Lysine N-Methyltransferase genetics, Smad3 Protein genetics, Transforming Growth Factor beta genetics
- Abstract
SMYD3 is a methylase previously linked to cancer cell invasion and migration. Here we show that SMYD3 favors TGFβ-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 TGFβ-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 TGFβ-dependent mesenchymal gene expression and cell migration in breast cancer, and support SMYD3 as a promising pharmacological target for anti-cancer therapy., (© The Author(s) 2018. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2019
- Full Text
- View/download PDF
19. The Trithorax protein Ash1L promotes myoblast fusion by activating Cdon expression.
- Author
-
Castiglioni I, Caccia R, Garcia-Manteiga JM, Ferri G, Caretti G, Molineris I, Nishioka K, and Gabellini D
- Subjects
- Animals, Cell Adhesion Molecules genetics, Cell Line, DNA-Binding Proteins, Histone-Lysine N-Methyltransferase genetics, Mice, Mice, Inbred C57BL, Muscular Dystrophies, Cell Adhesion Molecules metabolism, Histone-Lysine N-Methyltransferase metabolism, Muscle, Skeletal metabolism, Myoblasts metabolism
- Abstract
Myoblast fusion (MF) is required for muscle growth and repair, and its alteration contributes to muscle diseases. The mechanisms governing this process are incompletely understood, and no epigenetic regulator has been previously described. Ash1L is an epigenetic activator belonging to the Trithorax group of proteins and is involved in FSHD muscular dystrophy, autism and cancer. Its physiological role in skeletal muscle is unknown. Here we report that Ash1L expression is positively correlated with MF and reduced in Duchenne muscular dystrophy. In vivo, ex vivo and in vitro experiments support a selective and evolutionary conserved requirement for Ash1L in MF. RNA- and ChIP-sequencing indicate that Ash1L is required to counteract Polycomb repressive activity to allow activation of selected myogenesis genes, in particular the key MF gene Cdon. Our results promote Ash1L as an important epigenetic regulator of MF and suggest that its activity could be targeted to improve cell therapy for muscle diseases.
- Published
- 2018
- Full Text
- View/download PDF
20. Epigenetic targeting of bromodomain protein BRD4 counteracts cancer cachexia and prolongs survival.
- Author
-
Segatto M, Fittipaldi R, Pin F, Sartori R, Dae Ko K, Zare H, Fenizia C, Zanchettin G, Pierobon ES, Hatakeyama S, Sperti C, Merigliano S, Sandri M, Filippakopoulos P, Costelli P, Sartorelli V, and Caretti G
- Subjects
- 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, Cachexia prevention & control, Neoplasms, Experimental therapy, Nuclear Proteins antagonists & inhibitors, Nuclear Proteins genetics, Transcription Factors antagonists & inhibitors, Transcription Factors genetics
- 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.
- Published
- 2017
- Full Text
- View/download PDF
21. Vitamin D and VDR in cancer cachexia and muscle regeneration.
- Author
-
Camperi A, Pin F, Costamagna D, Penna F, Menduina ML, Aversa Z, Zimmers T, Verzaro R, Fittipaldi R, Caretti G, Baccino FM, Muscaritoli M, and Costelli P
- Subjects
- 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, Cachexia metabolism, Muscle, Skeletal metabolism, Receptors, Calcitriol metabolism, Vitamin D metabolism
- 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.
- Published
- 2017
- Full Text
- View/download PDF
22. Epigenetic Regulation Shapes the Stem Cells State.
- Author
-
Caretti G, Berghella L, Juan A, Latella L, and Ryall J
- Published
- 2016
- Full Text
- View/download PDF
23. A SMYD3 Small-Molecule Inhibitor Impairing Cancer Cell Growth.
- Author
-
Peserico A, Germani A, Sanese P, Barbosa AJ, Di Virgilio V, Fittipaldi R, Fabini E, Bertucci C, Varchi G, Moyer MP, Caretti G, Del Rio A, and Simone C
- Subjects
- 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, Cell Proliferation drug effects, Histone-Lysine N-Methyltransferase antagonists & inhibitors, Histone-Lysine N-Methyltransferase metabolism
- 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., (© 2015 Wiley Periodicals, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
24. The methyltransferase SMYD3 mediates the recruitment of transcriptional cofactors at the myostatin and c-Met genes and regulates skeletal muscle atrophy.
- Author
-
Proserpio V, Fittipaldi R, Ryall JG, Sartorelli V, and Caretti G
- Subjects
- 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, 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
- 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
25. Tackling skeletal muscle cells epigenome in the next-generation sequencing era.
- Author
-
Fittipaldi R and Caretti G
- 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.
- Published
- 2012
- Full Text
- View/download PDF
26. In vitro profiling of epigenetic modifications underlying heavy metal toxicity of tungsten-alloy and its components.
- Author
-
Verma R, Xu X, Jaiswal MK, Olsen C, Mears D, Caretti G, and Galdzicki Z
- Subjects
- Animals, Calcium Channels, L-Type physiology, Cells, Cultured, Chelating Agents pharmacology, Dose-Response Relationship, Drug, Egtazic Acid analogs & derivatives, Egtazic Acid pharmacology, Histones metabolism, Humans, Mice, Phosphorylation, Alloys toxicity, Epigenesis, Genetic, Tungsten toxicity
- Abstract
Tungsten-alloy has carcinogenic potential as demonstrated by cancer development in rats with intramuscular implanted tungsten-alloy pellets. This suggests a potential involvement of epigenetic events previously implicated as environmental triggers of cancer. Here, we tested metal induced cytotoxicity and epigenetic modifications including H3 acetylation, H3-Ser10 phosphorylation and H3-K4 trimethylation. We exposed human embryonic kidney (HEK293), human neuroepithelioma (SKNMC), and mouse myoblast (C2C12) cultures for 1-day and hippocampal primary neuronal cultures for 1-week to 50-200 μg/ml of tungsten-alloy (91% tungsten/6% nickel/3% cobalt), tungsten, nickel, and cobalt. We also examined the potential role of intracellular calcium in metal mediated histone modifications by addition of calcium channel blockers/chelators to the metal solutions. Tungsten and its alloy showed cytotoxicity at concentrations > 50 μg/ml, while we found significant toxicity with cobalt and nickel for most tested concentrations. Diverse cell-specific toxic effects were observed, with C2C12 being relatively resistant to tungsten-alloy mediated toxic impact. Tungsten-alloy, but not tungsten, caused almost complete dephosphorylation of H3-Ser10 in C2C12 and hippocampal primary neuronal cultures with H3-hypoacetylation in C2C12. Dramatic H3-Ser10 dephosphorylation was found in all cobalt treated cultures with a decrease in H3 pan-acetylation in C2C12, SKNMC and HEK293. Trimethylation of H3-K4 was not affected. Both tungsten-alloy and cobalt mediated H3-Ser10 dephosphorylation were reversed with BAPTA-AM, highlighting the role of intracellular calcium, confirmed with 2-photon calcium imaging. In summary, our results for the first time reveal epigenetic modifications triggered by tungsten-alloy exposure in C2C12 and hippocampal primary neuronal cultures suggesting the underlying synergistic effects of tungsten, nickel and cobalt mediated by changes in intracellular calcium homeostasis and buffering., (Published by Elsevier Inc.)
- Published
- 2011
- Full Text
- View/download PDF
27. Phosphoryl-EZH-ion.
- Author
-
Caretti G, Palacios D, Sartorelli V, and Puri PL
- Subjects
- Animals, Cell Cycle, Extracellular Signal-Regulated MAP Kinases metabolism, Humans, Models, Biological, Phosphorylation, Signal Transduction, Transcription Factors metabolism
- Abstract
Polycomb group (PcG) proteins regulate gene expression in embryonic and adult stem cells, but the mechanisms responsible for PcG gene targeting and regulation remain largely unknown. Recent evidence shows that EZH2, the enzymatic subunit of Polycomb Repressive Complex 2 (PRC2), is a nuclear phosphoprotein linking cell-cycle-intrinsic or extracellular signals to specific epigenetic signatures., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
28. TNF/p38α/polycomb signaling to Pax7 locus in satellite cells links inflammation to the epigenetic control of muscle regeneration.
- Author
-
Palacios D, Mozzetta C, Consalvi S, Caretti G, Saccone V, Proserpio V, Marquez VE, Valente S, Mai A, Forcales SV, Sartorelli V, and Puri PL
- Subjects
- Animals, Cells, Cultured, Epigenesis, Genetic, Fluorescent Antibody Technique, Gene Knockdown Techniques, Inflammation genetics, Mice, Mice, Inbred C57BL, PAX7 Transcription Factor genetics, Polycomb-Group Proteins, Promoter Regions, Genetic, Quadriceps Muscle metabolism, Satellite Cells, Skeletal Muscle cytology, Satellite Cells, Skeletal Muscle metabolism, PAX7 Transcription Factor metabolism, Quadriceps Muscle physiology, Regeneration, Repressor Proteins metabolism, Signal Transduction, Tumor Necrosis Factor-alpha metabolism, p38 Mitogen-Activated Protein Kinases metabolism
- Abstract
How regeneration cues are converted into the epigenetic information that controls gene expression in adult stem cells is currently unknown. We identified an inflammation-activated signaling in muscle stem (satellite) cells, by which the polycomb repressive complex 2 (PRC2) represses Pax7 expression during muscle regeneration. TNF-activated p38α kinase promotes the interaction between YY1 and PRC2, via threonine 372 phosphorylation of EZH2, the enzymatic subunit of the complex, leading to the formation of repressive chromatin on Pax7 promoter. TNF-α antibodies stimulate satellite cell proliferation in regenerating muscles of dystrophic or normal mice. Genetic knockdown or pharmacological inhibition of the enzymatic components of the p38/PRC2 signaling--p38α and EZH2--invariably promote Pax7 expression and expansion of satellite cells that retain their differentiation potential upon signaling resumption. Genetic knockdown of Pax7 impaired satellite cell proliferation in response to p38 inhibition, thereby establishing the biological link between p38/PRC2 signaling to Pax7 and satellite cell decision to proliferate or differentiate., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
29. Posttranslational regulation of NF-YA modulates NF-Y transcriptional activity.
- Author
-
Manni I, Caretti G, Artuso S, Gurtner A, Emiliozzi V, Sacchi A, Mantovani R, and Piaggio G
- Subjects
- Amino Acid Sequence, Animals, CCAAT-Binding Factor genetics, Cell Line, Humans, Lysine genetics, Lysine metabolism, Mice, Molecular Sequence Data, Promoter Regions, Genetic, Proteasome Endopeptidase Complex metabolism, Proteasome Inhibitors, Protein Subunits genetics, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Sequence Alignment, Ubiquitination, p300-CBP Transcription Factors metabolism, CCAAT-Binding Factor metabolism, Gene Expression Regulation, Protein Processing, Post-Translational, Protein Subunits metabolism, Transcription, Genetic
- Abstract
NF-Y binds to CCAAT motifs in the promoter region of a variety of genes involved in cell cycle progression. The NF-Y complex comprises three subunits, NF-YA, -YB, and -YC, all required for DNA binding. Expression of NF-YA fluctuates during the cell cycle and is down-regulated in postmitotic cells, indicating its role as the regulatory subunit of the complex. Control of NF-YA accumulation is posttranscriptional, NF-YA mRNA being relatively constant. Here we show that the levels of NF-YA protein are regulated posttranslationally by ubiquitylation and acetylation. A NF-YA protein carrying four mutated lysines in the C-terminal domain is more stable than the wild-type form, indicating that these lysines are ubiquitylated Two of the lysines are acetylated in vitro by p300, suggesting a competition between ubiquitylation and acetylation of overlapping residues. Interestingly, overexpression of a degradation-resistant NF-YA protein leads to sustained expression of mitotic cyclin complexes and increased cell proliferation, indicating that a tight regulation of NF-YA levels contributes to regulate NF-Y activity.
- Published
- 2008
- Full Text
- View/download PDF
30. p68 (Ddx5) interacts with Runx2 and regulates osteoblast differentiation.
- Author
-
Jensen ED, Niu L, Caretti G, Nicol SM, Teplyuk N, Stein GS, Sartorelli V, van Wijnen AJ, Fuller-Pace FV, and Westendorf JJ
- Subjects
- Animals, COS Cells, Cell Nucleus genetics, Cell Nucleus metabolism, Chlorocebus aethiops, Core Binding Factor Alpha 1 Subunit genetics, DEAD-box RNA Helicases genetics, Mice, Mice, Mutant Strains, Osteoblasts cytology, Protein Binding physiology, Skull cytology, Skull metabolism, Stem Cells cytology, Cell Differentiation physiology, Core Binding Factor Alpha 1 Subunit metabolism, DEAD-box RNA Helicases metabolism, Gene Expression Regulation physiology, Osteoblasts metabolism, Stem Cells metabolism
- 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., (2007 Wiley-Liss, Inc.)
- Published
- 2008
- Full Text
- View/download PDF
31. MyoD acetylation influences temporal patterns of skeletal muscle gene expression.
- Author
-
Di Padova M, Caretti G, Zhao P, Hoffman EP, and Sartorelli V
- Subjects
- Animals, Cell Lineage, Chromatin metabolism, Fibroblasts metabolism, Genome, Mice, Mice, Transgenic, Models, Genetic, Protein Processing, Post-Translational, RNA Polymerase II metabolism, Time Factors, Transcription, Genetic, Gene Expression Regulation, Developmental, Muscle, Skeletal metabolism, MyoD Protein metabolism, MyoD Protein physiology
- 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
32. The DEAD-box p68/p72 proteins and the noncoding RNA steroid receptor activator SRA: eclectic regulators of disparate biological functions.
- Author
-
Caretti G, Lei EP, and Sartorelli V
- Subjects
- Models, Biological, RNA, Long Noncoding, Cell Cycle physiology, DEAD-box RNA Helicases metabolism, Gene Expression Regulation, RNA, Untranslated metabolism, Regulatory Elements, Transcriptional physiology
- Abstract
p68 and p72 are RNA-binding proteins endowed with RNA helicase and RNA-protein complex remodeling activities. One of the RNAs associated with p68/p72 is the noncoding Steroid Receptors RNA Activator (SRA). Here we review recent findings on the cellular processes regulated by either p68/p72 alone or in combination with SRA and discuss the transcriptional events influenced by these molecules.
- Published
- 2007
- Full Text
- View/download PDF
33. The RNA helicases p68/p72 and the noncoding RNA SRA are coregulators of MyoD and skeletal muscle differentiation.
- Author
-
Caretti G, Schiltz RL, Dilworth FJ, Di Padova M, Zhao P, Ogryzko V, Fuller-Pace FV, Hoffman EP, Tapscott SJ, and Sartorelli V
- Subjects
- Amino Acid Sequence, HeLa Cells, Humans, Molecular Sequence Data, Muscle, Skeletal cytology, MyoD Protein genetics, RNA Helicases chemistry, Sequence Homology, Amino Acid, Cell Differentiation physiology, Muscle, Skeletal physiology, MyoD Protein metabolism, RNA Helicases metabolism, RNA, Untranslated metabolism
- Abstract
MyoD regulates skeletal myogenesis. Since proteins associated with MyoD exert regulatory functions, their identification is expected to contribute important insights into the mechanisms governing gene expression in skeletal muscle. We have found that the RNA helicases p68/p72 are MyoD-associated proteins and that the noncoding RNA SRA also immunoprecipitates with MyoD. In vitro and in vivo experiments indicated that both p68/p72 and SRA are coactivators of MyoD. RNA interference toward either p68/p72 or SRA prevented proper activation of muscle gene expression and cell differentiation. Unexpectedly, reducing the levels of p68/p72 proteins impaired recruitment of the TATA binding protein TBP; RNA polymerase II; and the catalytic subunit of the ATPase SWI/SNF complex, Brg-1, and hindered chromatin remodeling. These findings reveal that p68/p72 play a critical role in promoting the assembly of proteins required for the formation of the transcription initiation complex and chromatin remodeling.
- Published
- 2006
- Full Text
- View/download PDF
34. The Pole3 bidirectional unit is regulated by MYC and E2Fs.
- Author
-
Bolognese F, Forni C, Caretti G, Frontini M, Minuzzo M, and Mantovani R
- Subjects
- Animals, Chromatin Immunoprecipitation methods, DNA Polymerase II genetics, Histones biosynthesis, Histones genetics, Mice, Mutagenesis genetics, NIH 3T3 Cells, Point Mutation, Poly-ADP-Ribose Binding Proteins, Protein Structure, Tertiary genetics, Response Elements genetics, TATA Box genetics, DNA Polymerase II biosynthesis, Gene Expression Regulation physiology, S Phase physiology, Transcription, Genetic physiology
- Abstract
Pole3 (DPB4/YBL1/CHRAC17) is one of the subunits of the DNA polymerase e. It contains a histone-like domain required for the hererodimerization with its Pole4 (DPB3) partner. In another interaction, Pole3 heterodimerizes with YCL1/CHRAC15 and associates with the ACF1/SNF2H remodelling complex. We find that the Pol3 gene is regulated in starved NIH3T3 fibroblasts upon induction with serum, with a peak at the entry in the S phase. We characterized the Pole3 promoter, which is linked bidirectionally to C9Orf46, a gene of unknown function: it has no CCAAT nor TATA-boxes, and contains an E box and two potential E2F sites. Mutagenesis analysis points to a minimal promoter region as sufficient for activation; the E box and a neighbouring direct repeat are important for regulation. Cell-cycle regulation was reproduced in stable clones and an additional E2F site was found to be important. Chromatin immunoprecipitation analysis indicates that E2F1/4, as well as MYC, are associated with the Pole3 promoter in a phase-specific way. These data highlight coregulation of a histone-like gene with core histones upon DNA synthesis, and represent a first dissection of the interplay between two essential cell-cycle regulators on a bidirectional promoter.
- Published
- 2006
- Full Text
- View/download PDF
35. Fgfr4 is required for effective muscle regeneration in vivo. Delineation of a MyoD-Tead2-Fgfr4 transcriptional pathway.
- Author
-
Zhao P, Caretti G, Mitchell S, McKeehan WL, Boskey AL, Pachman LM, Sartorelli V, and Hoffman EP
- Subjects
- Animals, Cell Differentiation physiology, DNA-Binding Proteins genetics, Introns, Mice, Mice, Mutant Strains, Muscle, Skeletal pathology, Mutagenesis, MyoD Protein genetics, Myoblasts, Skeletal pathology, Myoblasts, Skeletal physiology, Promoter Regions, Genetic physiology, Receptor, Fibroblast Growth Factor, Type 4 genetics, TEA Domain Transcription Factors, Transcription Factors genetics, Transcription, Genetic physiology, Transfection, DNA-Binding Proteins metabolism, Muscle, Skeletal physiology, MyoD Protein metabolism, Receptor, Fibroblast Growth Factor, Type 4 metabolism, Regeneration physiology, Transcription Factors metabolism
- 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
36. Mechanisms underlying the transcriptional regulation of skeletal myogenesis.
- Author
-
Sartorelli V and Caretti G
- Subjects
- Animals, Basic Helix-Loop-Helix Transcription Factors, DNA-Binding Proteins metabolism, Humans, Muscle, Skeletal metabolism, Transcription Factors metabolism, Gene Expression Regulation, Developmental, Muscle, Skeletal embryology, Transcription, Genetic
- 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
37. The Polycomb Ezh2 methyltransferase regulates muscle gene expression and skeletal muscle differentiation.
- Author
-
Caretti G, Di Padova M, Micales B, Lyons GE, and Sartorelli V
- Subjects
- Animals, Cell Differentiation physiology, Chromatin metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Enhancer of Zeste Homolog 2 Protein, Erythroid-Specific DNA-Binding Factors, Extremities embryology, Histone Deacetylase 1, Histone Deacetylases genetics, Histone Deacetylases metabolism, Histone-Lysine N-Methyltransferase, Lysine metabolism, Methylation, Methyltransferases genetics, Methyltransferases metabolism, Mice, Mice, Inbred Strains, MyoD Protein genetics, MyoD Protein metabolism, Polycomb Repressive Complex 2, Protein Structure, Tertiary, Proteins genetics, RNA, Small Interfering, Regulatory Sequences, Nucleic Acid, Serum Response Factor genetics, Serum Response Factor metabolism, Transcription Factors genetics, Transcription Factors metabolism, Transcriptional Activation, YY1 Transcription Factor, Gene Expression Regulation, Developmental, Muscle, Skeletal cytology, Muscle, Skeletal physiology, Proteins metabolism
- 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
38. Deacetylase inhibitors increase muscle cell size by promoting myoblast recruitment and fusion through induction of follistatin.
- Author
-
Iezzi S, Di Padova M, Serra C, Caretti G, Simone C, Maklan E, Minetti G, Zhao P, Hoffman EP, Puri PL, and Sartorelli V
- Subjects
- Animals, Antibodies pharmacology, Cell Differentiation drug effects, Cell Differentiation genetics, Cyclic AMP Response Element-Binding Protein, DNA-Binding Proteins metabolism, Enzyme Inhibitors pharmacology, Enzyme Inhibitors therapeutic use, Female, Follistatin genetics, Follistatin metabolism, Histone Deacetylases metabolism, Humans, Hydroxamic Acids pharmacology, Membrane Fusion genetics, Mice, Muscle Fibers, Skeletal cytology, Muscle Fibers, Skeletal drug effects, Muscle, Skeletal cytology, Muscle, Skeletal enzymology, MyoD Protein metabolism, Myoblasts, Skeletal cytology, Myoblasts, Skeletal drug effects, NFATC Transcription Factors, NIH 3T3 Cells, RNA Interference, Regeneration drug effects, Regeneration genetics, Transcription Factors metabolism, Follistatin antagonists & inhibitors, Histone Deacetylase Inhibitors, Muscle Fibers, Skeletal enzymology, Muscle, Skeletal growth & development, Myoblasts, Skeletal enzymology, Nuclear Proteins
- Abstract
Fusion of undifferentiated myoblasts into multinucleated myotubes is a prerequisite for developmental myogenesis and postnatal muscle growth. We report that deacetylase inhibitors favor the recruitment and fusion of myoblasts into preformed myotubes. Muscle-restricted expression of follistatin is induced by deacetylase inhibitors and mediates myoblast recruitment and fusion into myotubes through a pathway distinct from those utilized by either IGF-1 or IL-4. Blockade of follistatin expression by RNAi-mediated knockdown, functional inactivation with either neutralizing antibodies or the antagonist protein myostatin, render myoblasts refractory to HDAC inhibitors. Muscles from animals treated with the HDAC inhibitor trichostatin A display increased production of follistatin and enhanced expression of markers of regeneration following muscle injury. These data identify follistatin as a central mediator of the fusigenic effects exerted by deacetylase inhibitors on skeletal muscles and establish a rationale for their use to manipulate skeletal myogenesis and promote muscle regeneration.
- Published
- 2004
- Full Text
- View/download PDF
39. Transcriptional activation of the cyclin A gene by the architectural transcription factor HMGA2.
- Author
-
Tessari MA, Gostissa M, Altamura S, Sgarra R, Rustighi A, Salvagno C, Caretti G, Imbriano C, Mantovani R, Del Sal G, Giancotti V, and Manfioletti G
- Subjects
- Adenovirus E4 Proteins chemistry, Adenovirus E4 Proteins genetics, Adenovirus E4 Proteins metabolism, Animals, Base Sequence, Binding Sites, CHO Cells, Cell Cycle, Cell Line, Cell Transformation, Neoplastic, Cricetinae, DNA, Complementary genetics, HMGA2 Protein genetics, Humans, Mice, Models, Biological, NIH 3T3 Cells, Promoter Regions, Genetic, Repressor Proteins chemistry, Repressor Proteins genetics, Repressor Proteins metabolism, Transcriptional Activation, Zinc Fingers, Cyclin A genetics, HMGA2 Protein metabolism
- Abstract
The HMGA2 protein belongs to the HMGA family of architectural transcription factors, which play an important role in chromatin organization. HMGA proteins are overexpressed in several experimental and human tumors and have been implicated in the process of neoplastic transformation. Hmga2 knockout results in the pygmy phenotype in mice and in a decreased growth rate of embryonic fibroblasts, thus indicating a role for HMGA2 in cell proliferation. Here we show that HMGA2 associates with the E1A-regulated transcriptional repressor p120(E4F), interfering with p120(E4F) binding to the cyclin A promoter. Ectopic expression of HMGA2 results in the activation of the cyclin A promoter and induction of the endogenous cyclin A gene. In addition, chromatin immunoprecipitation experiments show that HMGA2 associates with the cyclin A promoter only when the gene is transcriptionally activated. These data identify the cyclin A gene as a cellular target for HMGA2 and, for the first time, suggest a mechanism for HMGA2-dependent cell cycle regulation.
- Published
- 2003
- Full Text
- View/download PDF
40. Dynamic recruitment of NF-Y and histone acetyltransferases on cell-cycle promoters.
- Author
-
Caretti G, Salsi V, Vecchi C, Imbriano C, and Mantovani R
- Subjects
- 3T3 Cells, Animals, Cell Nucleus enzymology, DNA-Binding Proteins metabolism, E2F4 Transcription Factor, E2F6 Transcription Factor, G2 Phase physiology, Histone Acetyltransferases, Histones metabolism, Mice, Mitosis physiology, Protein Binding physiology, S Phase physiology, Transcription Factors metabolism, Acetyltransferases metabolism, CCAAT-Binding Factor metabolism, G1 Phase physiology, Promoter Regions, Genetic physiology, Saccharomyces cerevisiae Proteins metabolism
- Abstract
Regulation of transcription during the cell-cycle is under the control of E2 factors (E2Fs), often in cooperation with nuclear factor Y (NF-Y), a histone-like CCAAT-binding trimer. NF-Y is paradigmatic of a constitutive, ubiquitous factor that pre-sets the promoter architecture for other regulatory proteins to access it. We analyzed the recruitment of NF-Y, E2F1/4/6, histone acetyltransferases, and histone deacetylase (HDAC) 1/3/4 to several cell-cycle promoters by chromatin immunoprecipitation assays in serum-starved and restimulated NIH3T3 cells. NF-Y binding is not constitutive but timely regulated in all promoters tested, being displaced when promoters are repressed. p300 association correlates with activation, and it is never found in the absence of NF-Y, whereas PCAF/hGCN5 is often found before NF-Y association. E2F4 and E2F6, together with HDACs, are bound to repressed promoters, including the G2/M Cyclin B2. As expected, an inverse relationship between HDACs association and histones H3/H4 acetylation is observed. Blocking cells in G1 with the cyclin-dependent kinase 2 inhibitor R-roscovitine confirms that NF-Y is bound to G1/S but not to G2/M promoters in G1. These data indicate that following the release of E2Fs/HDACs, a hierarchy of PCAF-NF-Y-p300 interactions and H3-H4 acetylations are required for activation of cell-cycle promoters.
- Published
- 2003
- Full Text
- View/download PDF
41. Interactions between p300 and multiple NF-Y trimers govern cyclin B2 promoter function.
- Author
-
Salsi V, Caretti G, Wasner M, Reinhard W, Haugwitz U, Engeland K, and Mantovani R
- Subjects
- 3T3 Cells, Animals, Base Sequence, Cell Cycle, Cell Line, Chromatin metabolism, Cyclin B chemistry, Cyclin B2, Dimerization, E1A-Associated p300 Protein, Escherichia coli metabolism, Humans, Insecta, Luciferases metabolism, Mice, Models, Biological, Molecular Sequence Data, Mutation, Phosphorylation, Plasmids metabolism, Precipitin Tests, Promoter Regions, Genetic, Protein Binding, Transcriptional Activation, Transfection, Tumor Cells, Cultured, CCAAT-Binding Factor metabolism, Cyclin B genetics, Nuclear Proteins metabolism, Trans-Activators metabolism
- Abstract
The CCAAT box is one of the most common elements in eukaryotic promoters and is activated by NF-Y, a conserved trimeric transcription factor with histone-like subunits. Usually one CCAAT element is present in promoters at positions between -60 and -100, but an emerging class of promoters harbor multiple NF-Y sites. In the triple CCAAT-containing cyclin B2 cell-cycle promoter, all CCAAT boxes, independently from their NF-Y affinities, are important for function. We investigated the relationships between NF-Y and p300. Chromatin immunoprecipitation analysis found that NF-Y and p300 are bound to the cyclin B2 promoter in vivo and that their binding is regulated during the cell cycle, positively correlating with promoter function. Cotransfection experiments determined that the coactivator acts on all CCAAT boxes and requires a precise spacing between the three elements. We established the order of in vitro binding of the three NF-Y complexes and find decreasing affinities from the most distal Y1 to the proximal Y3 site. Binding of two or three NF-Y trimers with or without p300 is not cooperative, but association with the Y1 and Y2 sites is extremely stable. p300 favors the binding of NF-Y to the weak Y3 proximal site, provided that a correct distance between the three CCAAT is respected. Our data indicate that the precise spacing of multiple CCAAT boxes is crucial for coactivator function. Transient association to a weak site might be a point of regulation during the cell cycle and a general theme of multiple CCAAT box promoters.
- Published
- 2003
- Full Text
- View/download PDF
42. Cloning and characterization of the histone-fold proteins YBL1 and YCL1.
- Author
-
Bolognese F, Imbriano C, Caretti G, and Mantovani R
- Subjects
- Algorithms, Amino Acid Motifs, Amino Acid Sequence, Animals, CCAAT-Binding Factor chemistry, CCAAT-Binding Factor metabolism, Cell Line, Centrifugation, Density Gradient, Cloning, Molecular, DNA chemistry, DNA genetics, DNA metabolism, DNA-Binding Proteins chemistry, Dimerization, Embryo, Mammalian metabolism, Expressed Sequence Tags, Histones metabolism, Humans, Macromolecular Substances, Mice, Molecular Sequence Data, Molecular Weight, Nucleosomes chemistry, Nucleosomes genetics, Phosphoproteins chemistry, Phosphoproteins metabolism, Phylogeny, Protein Binding, RNA, Messenger analysis, RNA, Messenger genetics, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Response Elements genetics, Sequence Alignment, Solutions, Structure-Activity Relationship, TATA Box genetics, Transcription Factors chemistry, Transcription Factors metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Histones chemistry, Histones genetics, Nucleosomes metabolism
- Abstract
Histones are among the most conserved proteins in evolution, sharing a histone fold motif. A number of additional histonic proteins exist and are involved in the process of transcriptional regulation. We describe here the identification, cloning and characterization of two small members of the H2A-H2B sub-family (YBL1 and YCL1) related to the NF-YB and NF-YC subunits of the CCAAT-binding activator NF-Y and to the TATA-binding protein (TBP) binding repressor NC2. Unlike the latters, YBL1 and YCL1 have no intrinsic CCAAT or TATA-binding capacity. In nucleosome reconstitution assays, they can form complexes with histones in solution and on DNA and they are part of relatively large complexes, as determined by glycerol gradient experiments. Our data support the idea that YBL1 and YCL1 are divergent with respect to NF-YB and NF-YC for specific functions, but have coevolved the capacity to interact with nucleosomal structures.
- Published
- 2000
- Full Text
- View/download PDF
43. Dissection of functional NF-Y-RFX cooperative interactions on the MHC class II Ea promoter.
- Author
-
Caretti G, Cocchiarella F, Sidoli C, Villard J, Peretti M, Reith W, and Mantovani R
- Subjects
- Animals, Binding Sites, CCAAT-Enhancer-Binding Proteins, Cell Line, Conserved Sequence genetics, DNA chemistry, DNA genetics, DNA metabolism, DNA-Binding Proteins genetics, Kinetics, Mice, Mutation, Nucleic Acid Conformation, Nucleosomes chemistry, Nucleosomes genetics, Nucleosomes metabolism, Precipitin Tests, Protein Binding, Recombinant Proteins metabolism, Regulatory Factor X Transcription Factors, Response Elements genetics, Thermodynamics, Trans-Activators genetics, Trans-Activators metabolism, Transcription Factors genetics, Transcription, Genetic genetics, Transcriptional Activation genetics, Transfection, DNA-Binding Proteins metabolism, Genes, MHC Class II genetics, Nuclear Proteins, Promoter Regions, Genetic genetics, Transcription Factors metabolism
- Abstract
Transcription of major histocompatibility complex (MHC) class II genes depends upon the trimeric complexes RFX and NF-Y binding to the conserved X-Y promoter elements. We produced and purified the RFX subunits from Escherichia coli, reconstituted DNA-binding to the mouse Ea X box and dissected the interactions with NF-Y. RFX and NF-Y do not interact in solution, but make cooperative interactions in EMSA: a minimal NF-Y, composed of the evolutionary conserved domains, is sufficient and the RFXAP N-terminal half is expendable. Altering the X-Y distance abolishes cooperativity, indicating that DNA imposes severe spatial constraints. When tested on a highly positioned nucleosome, RFX binds DNA well and NF-Y does not increase its affinity further. Transfections of NF-Y subunits, but not RFX, in class II negative cells improves basal transcription and coexpression of the two activators has a synergistic effect, while modestly increasing CIITA-mediated activation. These results show that interactions between the two trimers on DNA are key to MHC class II expression., (Copyright 2000 Academic Press.)
- Published
- 2000
- Full Text
- View/download PDF
44. A functionally essential domain of RFX5 mediates activation of major histocompatibility complex class II promoters by promoting cooperative binding between RFX and NF-Y.
- Author
-
Villard J, Peretti M, Masternak K, Barras E, Caretti G, Mantovani R, and Reith W
- Subjects
- Amino Acid Sequence, Animals, B-Lymphocytes, CCAAT-Enhancer-Binding Proteins, Conserved Sequence, DNA-Binding Proteins genetics, Genetic Complementation Test, Humans, Mice, Mice, Inbred BALB C, Molecular Sequence Data, Peptide Fragments genetics, Peptide Fragments metabolism, Protein Binding, Protein Structure, Tertiary, Regulatory Factor X Transcription Factors, Sequence Homology, Amino Acid, Transcriptional Activation, DNA-Binding Proteins metabolism, Histocompatibility Antigens Class II genetics, Major Histocompatibility Complex genetics, Promoter Regions, Genetic, Transcription Factors metabolism
- Abstract
Major histocompatibility complex class II (MHC-II) molecules occupy a pivotal position in the adaptive immune system, and correct regulation of their expression is therefore of critical importance for the control of the immune response. Several regulatory factors essential for the transcription of MHC-II genes have been identified by elucidation of the molecular defects responsible for MHC-II deficiency, a hereditary immunodeficiency disease characterized by regulatory defects abrogating MHC-II expression. Three of these factors, RFX5, RFXAP, and RFXANK, combine to form the RFX complex, a regulatory protein that binds to the X box DNA sequence present in all MHC-II promoters. In this study we have undertaken a dissection of the structure and function of RFX5, the largest subunit of the RFX complex. The results define two distinct domains serving two different essential functions. A highly conserved N-terminal region of RFX5 is required for its association with RFXANK and RFXAP, for assembly of the RFX complex in vivo and in vitro, and for binding of this complex to its X box target site in the MHC-II promoter. This N-terminal region is, however, not sufficient for activation of MHC-II expression. This requires an additional domain within the C-terminal region of RFX5. This C-terminal domain mediates cooperative binding between the RFX complex and NF-Y, a transcription factor binding to the Y box sequence of MHC-II promoters. This provides direct evidence that RFX5-mediated cooperative binding between RFX and NF-Y plays an essential role in the transcriptional activation of MHC-II genes.
- Published
- 2000
- Full Text
- View/download PDF
45. NF-Y associates with H3-H4 tetramers and octamers by multiple mechanisms.
- Author
-
Caretti G, Motta MC, and Mantovani R
- Subjects
- Animals, Artemia, Base Sequence, CCAAT-Enhancer-Binding Proteins, Chickens, DNA metabolism, DNA-Binding Proteins genetics, Deoxyribonuclease I metabolism, Dimerization, Exodeoxyribonucleases metabolism, Molecular Sequence Data, Nucleosomes metabolism, Protein Binding, Solutions, Xenopus laevis, DNA-Binding Proteins metabolism, Histones metabolism
- Abstract
NF-Y is a CCAAT-binding trimer with two histonic subunits, NF-YB and NF-YC, resembling H2A-H2B. We previously showed that the short conserved domains of NF-Y efficiently bind to the major histocompatibility complex class II Ea Y box in DNA nucleosomized with purified chicken histones. Using wild-type NF-Y and recombinant histones, we find that NF-Y associates with H3-H4 early during nucleosome assembly, under conditions in which binding to naked DNA is not observed. In such assays, the NF-YB-NF-YC dimer forms complexes with H3-H4, for whose formation the CCAAT box is not required. We investigated whether they represent octamer-like structures, using DNase I, micrococcal nuclease, and exonuclease III, and found a highly positioned nucleosome on Ea, whose boundaries were mapped; addition of NF-YB-NF-YC does not lead to the formation of octameric structures, but changes in the digestion patterns are observed. NF-YA can bind to such preformed DNA complexes in a CCAAT-dependent way. In the absence of DNA, NF-YB-NF-YC subunits bind to H3-H4, but not to H2A-H2B, through the NF-YB histone fold. These results indicate that (i) the NF-Y histone fold dimer can efficiently associate DNA during nucleosome formation; (ii) it has an intrinsic affinity for H3-H4 but does not form octamers; and (iii) the interactions between NF-YA, NF-YB-NF-YC, and H3-H4 or nucleosomes are not mutually exclusive. Thus, NF-Y can intervene at different steps during nucleosome formation, and this scenario might be paradigmatic for other histone fold proteins involved in gene regulation.
- Published
- 1999
- Full Text
- View/download PDF
46. Effect of ecteinascidin-743 on the interaction between DNA binding proteins and DNA.
- Author
-
Bonfanti M, La Valle E, Fernandez Sousa Faro JM, Faircloth G, Caretti G, Mantovani R, and D'Incalci M
- Subjects
- Animals, Base Sequence, CCAAT-Enhancer-Binding Proteins, Consensus Sequence, DNA-Binding Proteins antagonists & inhibitors, Electrophoresis, Leukemia L1210 metabolism, Mice, Oligonucleotides antagonists & inhibitors, Oligonucleotides metabolism, Proto-Oncogene Proteins antagonists & inhibitors, Proto-Oncogene Proteins metabolism, Sp1 Transcription Factor antagonists & inhibitors, Sp1 Transcription Factor metabolism, TATA-Box Binding Protein, Tetrahydroisoquinolines, Trabectedin, Transcription Factors antagonists & inhibitors, Antineoplastic Agents, Alkylating pharmacology, DNA metabolism, DNA-Binding Proteins metabolism, Dioxoles pharmacology, Isoquinolines pharmacology, Transcription Factors metabolism
- Abstract
Ecteinascidin-743 (ET-743) is a tetrahydroisoquinoline alkaloid isolated from Ecteinascidia turbinata, a tunicate growing in mangrove roots in Caribbean. It has been shown to bind in the minor groove of DNA forming covalent adducts by reaction of the N2 of guanine with the carbinolamine moiety. We investigated ET-743 ability to inhibit the binding of different transcription factors to their consensus sequences by using gel shift assays. We have selected three types of factors: (i) oncogene products such as MYC, c-MYB and Maf; (ii) transcriptional activators regulated during the cell cycle as E2F and SRF; and (iii) general transcription factors such as TATA binding protein (TBP), Sp1 and NF-Y. We observed no inhibition of the binding of Sp1, Maf, MYB and MYC. Inhibition of DNA binding was observed for TBP, E2F, SRF at ET-743 concentrations ranging from 50 to 300 microM. The inhibition of binding of NF-Y occurs at even lower concentrations (i.e. 10-30 microM) when the recombinant subunits of NF-Y are preincubated with the drug, indicating that the inhibition of NF-Y binding does not require previous ET-743 DNA binding. Since NF-Y is a trimer containing two subunits with high resemblance to histones H2B and H2A, we have investigated the effect of ET-743 on nucleosome reconstitution. ET-743 caused a decrease of the nucleosomal band at 100 nM, with the complete disappearance of the band at 3-10 microM. These data suggest that the mode of action of this novel anticancer drug is related to its ability to modify the interaction between some DNA binding proteins and DNA.
- Published
- 1999
47. In vivo analysis of the state of the human uPA enhancer following stimulation by TPA.
- Author
-
Ibañez-Tallon I, Caretti G, Blasi F, and Crippa MP
- Subjects
- Binding Sites, Chromatin genetics, Chromatin ultrastructure, Deoxyribonuclease I genetics, Deoxyribonuclease I metabolism, Humans, Micrococcal Nuclease metabolism, NF-kappa B metabolism, Nucleosomes genetics, Nucleosomes metabolism, RNA, Messenger drug effects, Regulatory Sequences, Nucleic Acid, Restriction Mapping, Time Factors, Transcription Factors metabolism, Transcription, Genetic, Urokinase-Type Plasminogen Activator drug effects, Carcinogens pharmacology, Enhancer Elements, Genetic, Tetradecanoylphorbol Acetate pharmacology, Urokinase-Type Plasminogen Activator genetics, Urokinase-Type Plasminogen Activator metabolism
- Abstract
We have analysed in vivo the -2.0 kb enhancer of the human urokinase-type plasminogen activator (uPA) gene in HepG2 cells, in which gene expression can be induced by phorbol esters. The results reveal that, within the regulatory region, the enhancer, the silencer and the minimal promoter become hypersensitive to deoxyribonuclease I (DNase I) upon induction of transcription. The hypersensitivity of the enhancer can be reversed after removal of the inducer. In vivo footprinting analysis indicates that all the cis-acting elements of the enhancer, previously identified in vitro, are occupied in vivo upon 12-O-tetradecanoyl-phorbol-13-acetate (TPA) stimulation of HepG2 cells. Micrococcal nuclease (MNase) cleavage of this region fails to reveal discrete nucleosomal boundaries in vivo in close proximity of the enhancer, either before or after stimulation. Furthermore, this region does not lose its nucleosomal configuration after TPA induction of transcription. An approximately 600 bp long region around the enhancer becomes more, but not fully, accessible to restriction endonucleases upon stimulation. A time-course experiment shows that this accessibility reaches a plateau after a 1 h TPA treatment suggesting the persistent presence of nucleosomes. These results indicate that TPA induces the binding of transcription factors to the uPA enhancer without chromatin remodelling of this region.
- Published
- 1999
- Full Text
- View/download PDF
48. Interactions of the CCAAT-binding trimer NF-Y with nucleosomes.
- Author
-
Motta MC, Caretti G, Badaracco GF, and Mantovani R
- Subjects
- Animals, Artemia, CCAAT-Enhancer-Binding Proteins, Chromatin metabolism, DNA metabolism, DNA Footprinting, Histones metabolism, Male, Nucleosomes genetics, Oligonucleotides metabolism, Plasmids, Promoter Regions, Genetic, Protein Conformation, Protein Folding, Salmon, Spermatozoa chemistry, DNA-Binding Proteins metabolism, Nucleosomes metabolism, Transcription Factors metabolism
- Abstract
NF-Y is a sequence-specific evolutionary conserved activator binding to CCAAT boxes with high affinity and specificity. It is a trimer formed by NF-YA and two putative histone-like subunits, NF-YB and NF-YC, showing similarity to histones H2B and H2A, respectively. We investigated the relationships between NF-Y and chromatin using an Artemia franciscana chromatin assembly system with plasmids containing the Major HistoCompatibility complex class II Ea promoter. The NF-Y trimer, but not single subunits, protects the Y box in the presence of reconstituted chromatin, and it can bind the target sequence during and after assembly. Using reconstitution assays with purified chicken histones, we show that NF-Y associates with preformed nucleosomes. Translational analysis of various Ea fragments of identical length in which the CCAAT box is at different positions indicated that the lateral fragment was slightly more prone to NF-Y binding. In competition experiments, NF-Y is able to prevent formation of nucleosomes significantly. These data support the idea that NF-Y is a gene-specific activator with a built-in capacity to interface with chromatin structures.
- Published
- 1999
- Full Text
- View/download PDF
49. [Technical and physico-dosimetric comparisons between a telecobaltotherapy unit and a Van de Braaff electrostatic generator].
- Author
-
MICELI R, CORINALDESI A, and CARETTI G
- Subjects
- Humans, Cobalt, Cobalt Radioisotopes, Equipment and Supplies, Radioactivity, Radiometry, Radiotherapy supply & distribution, Static Electricity
- Published
- 1961
50. [SCATTERED RADIATION IN DIAGNOSIS AND THERAPY AS RELATED TO THE PROFESSIONAL RISK].
- Author
-
CARETTI G and CHIANURA G
- Subjects
- Humans, Radiation Protection, Radiography, Radiotherapy, Radiotherapy Dosage
- Published
- 1963
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.