53 results on '"Mariangela Morlando"'
Search Results
2. A longitudinal study defined circulating microRNAs as reliable biomarkers for disease prognosis and progression in ALS human patients
- Author
-
Gabriella Dobrowolny, Julie Martone, Elisa Lepore, Irene Casola, Antonio Petrucci, Maurizio Inghilleri, Mariangela Morlando, Alessio Colantoni, Bianca Maria Scicchitano, Andrea Calvo, Giulia Bisogni, Adriano Chiò, Mario Sabatelli, Irene Bozzoni, and Antonio Musarò
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Cytology ,QH573-671 - Abstract
Abstract Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease associated with motor neuron degeneration, muscle atrophy and paralysis. To date, multiple panels of biomarkers have been described in ALS patients and murine models. Nevertheless, none of them has sufficient specificity and thus the molecular signature for ALS prognosis and progression remains to be elucidated. Here we overcome this limitation through a longitudinal study, analyzing serum levels of circulating miRNAs, stable molecules that are recently used as promising biomarkers for many types of human disorders, in ALS patients during the progression of the pathology. We performed next-generation sequencing (NGS) analysis and absolute RT quantification of serum samples of ALS patients and healthy controls. The expression levels of five selected miRNAs were quantitatively analyzed during disease progression in each patient and we demonstrated that high levels of miR-206, miR-133a and miR-151a-5p can predict a slower clinical decline of patient functionality. In particular, we found that miR-206 and miR-151a-5p serum levels were significantly up-regulated at the mild stage of ALS pathology, to decrease in the following moderate and severe stages, whereas the expression levels of miR-133a and miR-199a-5p remained low throughout the course of the disease, showing a diagnostic significance in moderate and severe stages for miR-133a and in mild and terminal ones for miR-199a-5p. Moreover, we found that miR-423–3p and 151a-5p were significantly downregulated respectively in mild and terminal stages of the disease. These data suggest that these miRNAs represent potential prognostic markers for ALS disease.
- Published
- 2021
- Full Text
- View/download PDF
3. Circ-Hdgfrp3 shuttles along neurites and is trapped in aggregates formed by ALS-associated mutant FUS
- Author
-
Eleonora D'Ambra, Tiziana Santini, Erika Vitiello, Sara D'Uva, Valentina Silenzi, Mariangela Morlando, and Irene Bozzoni
- Subjects
Biological sciences ,Molecular neuroscience ,Cellular neuroscience ,Science - Abstract
Summary: CircRNAs belong to a family of RNA molecules which are conserved in evolution, have tissue-specific expression, and are abundant in neuronal cells. Here, we define several features of circ-Hdgfrp3 and describe interesting alterations occurring in motor neurons (MNs) carrying ALS-associated FUS mutations. Through a highly sensitive in situ approach we describe that circ-Hdgfrp3 traffics along neurites, while upon oxidative stress it is retained in the perinuclear region. While in wild-type stressed MNs, circ-Hdgfrp3 localizes in stress granules (SGs), in MNs carrying mutant FUS, a higher proportion of circ-Hdgfrp3 was trapped into cytoplasmic aggregates. Upon stress removal, circ-Hdgfrp3 was easily freed from SGs whereas it was less efficiently released from FUS-aggregates. We found that the human circ-Hdgfrp3 counterpart was also similarly associated to mutant FUS-aggregates in stressed neuronal cells. Overall, the alteration of circ-Hdgfrp3 trafficking adds a further layer of complexity to the role of FUS-aggregates in ALS disease.
- Published
- 2021
- Full Text
- View/download PDF
4. From Serendipity to Rational Identification of the 5,6,7,8-Tetrahydrobenzo[4,5]thieno[2,3-d]pyrimidin-4(3H)-one Core as a New Chemotype of AKT1 Inhibitors for Acute Myeloid Leukemia
- Author
-
Andrea Astolfi, Francesca Milano, Deborah Palazzotti, Jose Brea, Maria Chiara Pismataro, Mariangela Morlando, Oriana Tabarrini, Maria Isabel Loza, Serena Massari, Maria Paola Martelli, and Maria Letizia Barreca
- Subjects
AKT1 ,kinase inhibitors ,cancer ,acute myeloid leukemia ,rare disease ,molecular modeling ,Pharmacy and materia medica ,RS1-441 - Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematopoietic malignancy whose prognosis is globally poor. In more than 60% of AML patients, the PI3K/AKTs/mTOR signaling pathway is aberrantly activated because of oncogenic driver alterations and further enhanced by chemotherapy as a mechanism of drug resistance. Against this backdrop, very recently we have started a multidisciplinary research project focused on AKT1 as a pharmacological target to identify novel anti-AML agents. Indeed, the serendipitous finding of the in-house compound T187 as an AKT1 inhibitor has paved the way to the rational identification of new active small molecules, among which T126 has emerged as the most interesting compound with IC50 = 1.99 ± 0.11 μM, ligand efficiency of 0.35, and a clear effect at low micromolar concentrations on growth inhibition and induction of apoptosis in AML cells. The collected results together with preliminary SAR data strongly indicate that the 5,6,7,8-tetrahydrobenzo[4,5]thieno[2,3-d]pyrimidin-4(3H)-one derivative T126 is worthy of future biological experiments and medicinal chemistry efforts aimed at developing a novel chemical class of AKT1 inhibitors as anti-AML agents.
- Published
- 2022
- Full Text
- View/download PDF
5. Non-coding RNAs in Nervous System Development and Disease
- Author
-
Beatrice Salvatori, Silvia Biscarini, and Mariangela Morlando
- Subjects
ncRNAs ,circRNAs ,neurodegenerative diseases ,biomarkers ,therapeutics ,neuronal development ,Biology (General) ,QH301-705.5 - Abstract
The rapid advance of RNA sequencing technologies contributed to a deep understanding of transcriptome composition and has allowed the discovery of a large number of non-coding RNAs (ncRNAs). The ability of these RNA molecules to be engaged in intricate and dynamic interactions with proteins and nucleic acids led to a great expansion of gene expression regulation mechanisms. By this matter, ncRNAs contribute to the increase in regulatory complexity that becomes highly specific between tissues and cell types. Among the ncRNAs, long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) are especially abundant in nervous system and have been shown to be implicated in its development, plasticity and aging as well as in neurological disorders. This review provides an overview of how these two diverse classes of ncRNAs control cellular processes during nervous system development, physiology, and disease conditions with particular emphasis on neurodegenerative disorders. The use of ncRNAs as biomarkers, tools, or targets for therapeutic intervention in neurodegeneration are also discussed.
- Published
- 2020
- Full Text
- View/download PDF
6. The Long Non-coding RNA lnc-31 Interacts with Rock1 mRNA and Mediates Its YB-1-Dependent Translation
- Author
-
Dacia Dimartino, Alessio Colantoni, Monica Ballarino, Julie Martone, Davide Mariani, Johannes Danner, Astrid Bruckmann, Gunter Meister, Mariangela Morlando, and Irene Bozzoni
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Cytoplasmic long non-coding RNAs have been shown to act at many different levels to control post-transcriptional gene expression, although their role in translational control is poorly understood. Here, we show that lnc-31, a non-coding RNA required for myoblast proliferation, promotes ROCK1 protein synthesis by stabilizing its translational activator, YB-1. We find that lnc-31 binds to the Rock1 mRNA as well as to the YB-1 protein and that translational activation requires physical interaction between the two RNA species. These results suggest a localized effect of YB-1 stabilization on the Rock1 mRNA. ROCK1 upregulation by lnc-31, in proliferative conditions, correlates well with the differentiation-repressing activity of ROCK1. We also show that, upon induction of differentiation, the downregulation of lnc-31, in conjunction with miR-152 targeting of Rock1, establishes a regulatory loop that reinforces ROCK1 repression and promotes myogenesis. : Dimartino et al. demonstrate that lnc-31 is required to sustain myoblast proliferation. lnc-31 interacts with Rock1 mRNA, an inhibitor of differentiation, and promotes its translation. This activity is strengthened by binding of the translational regulator YB-1 and its lnc-31-dependent stabilization. Keywords: long non-coding RNA, myogenesis, Rock1, YB-1, lnc-31, cell cycle, translation, proteasome, protein stability
- Published
- 2018
- Full Text
- View/download PDF
7. FUS affects circular RNA expression in murine embryonic stem cell-derived motor neurons
- Author
-
Lorenzo Errichelli, Stefano Dini Modigliani, Pietro Laneve, Alessio Colantoni, Ivano Legnini, Davide Capauto, Alessandro Rosa, Riccardo De Santis, Rebecca Scarfò, Giovanna Peruzzi, Lei Lu, Elisa Caffarelli, Neil A. Shneider, Mariangela Morlando, and Irene Bozzoni
- Subjects
Science - Abstract
The RNA binding protein FUS functions in several RNA biosynthetic processes and has been linked to the pathogenesis of amyotrophic lateral sclerosis (ALS). Here the authors show that FUS controls back-splicing reactions leading to circular RNA (circRNA) production in stem cell-derived motor neurons and that ALS-associated FUS mutations affect the biogenesis of circRNAs.
- Published
- 2017
- Full Text
- View/download PDF
8. ALS mutant FUS proteins are recruited into stress granules in induced pluripotent stem cell-derived motoneurons
- Author
-
Jessica Lenzi, Riccardo De Santis, Valeria de Turris, Mariangela Morlando, Pietro Laneve, Andrea Calvo, Virginia Caliendo, Adriano Chiò, Alessandro Rosa, and Irene Bozzoni
- Subjects
ALS ,FUS ,TALE nucleases ,iPSCs ,Medicine ,Pathology ,RB1-214 - Abstract
Patient-derived induced pluripotent stem cells (iPSCs) provide an opportunity to study human diseases mainly in those cases for which no suitable model systems are available. Here, we have taken advantage of in vitro iPSCs derived from patients affected by amyotrophic lateral sclerosis (ALS) and carrying mutations in the RNA-binding protein FUS to study the cellular behavior of the mutant proteins in the appropriate genetic background. Moreover, the ability to differentiate iPSCs into spinal cord neural cells provides an in vitro model mimicking the physiological conditions. iPSCs were derived from FUSR514S and FUSR521C patient fibroblasts, whereas in the case of the severe FUSP525L mutation, in which fibroblasts were not available, a heterozygous and a homozygous iPSC line were raised by TALEN-directed mutagenesis. We show that aberrant localization and recruitment of FUS into stress granules (SGs) is a prerogative of the FUS mutant proteins and occurs only upon induction of stress in both undifferentiated iPSCs and spinal cord neural cells. Moreover, we show that the incorporation into SGs is proportional to the amount of cytoplasmic FUS, strongly correlating with the cytoplasmic delocalization phenotype of the different mutants. Therefore, the available iPSCs represent a very powerful system for understanding the correlation between FUS mutations, the molecular mechanisms of SG formation and ALS ethiopathogenesis.
- Published
- 2015
- Full Text
- View/download PDF
9. FUS Alters circRNA Metabolism in Human Motor Neurons Carrying the ALS-Linked P525L Mutation
- Author
-
Alessio Colantoni, Davide Capauto, Vincenzo Alfano, Eleonora D’Ambra, Sara D’Uva, Gian Gaetano Tartaglia, and Mariangela Morlando
- Subjects
FUS ,circRNAs ,P525L ,ALS ,back-splicing ,motor neurons ,ceRNA ,Alu ,ADAR ,Organic Chemistry ,General Medicine ,Catalysis ,Computer Science Applications ,Inorganic Chemistry ,Physical and Theoretical Chemistry ,Molecular Biology ,Spectroscopy - Abstract
Deregulation of RNA metabolism has emerged as one of the key events leading to the degeneration of motor neurons (MNs) in Amyotrophic Lateral Sclerosis (ALS) disease. Indeed, mutations on RNA-binding proteins (RBPs) or on proteins involved in aspects of RNA metabolism account for the majority of familiar forms of ALS. In particular, the impact of the ALS-linked mutations of the RBP FUS on many aspects of RNA-related processes has been vastly investigated. FUS plays a pivotal role in splicing regulation and its mutations severely alter the exon composition of transcripts coding for proteins involved in neurogenesis, axon guidance, and synaptic activity. In this study, by using in vitro-derived human MNs, we investigate the effect of the P525L FUS mutation on non-canonical splicing events that leads to the formation of circular RNAs (circRNAs). We observed altered levels of circRNAs in FUSP525L MNs and a preferential binding of the mutant protein to introns flanking downregulated circRNAs and containing inverted Alu repeats. For a subset of circRNAs, FUSP525L also impacts their nuclear/cytoplasmic partitioning, confirming its involvement in different processes of RNA metabolism. Finally, we assess the potential of cytoplasmic circRNAs to act as miRNA sponges, with possible implications in ALS pathogenesis.
- Published
- 2023
10. Best practice standards for circular RNA research
- Author
-
Anne F. Nielsen, Albrecht Bindereif, Irene Bozzoni, Mor Hanan, Thomas B. Hansen, Manuel Irimia, Sebastian Kadener, Lasse S. Kristensen, Ivano Legnini, Mariangela Morlando, Morten T. Jarlstad Olesen, R. Jeroen Pasterkamp, Stephan Preibisch, Nikolaus Rajewsky, Christin Suenkel, and Jørgen Kjems
- Subjects
splicing ,RNA Splicing ,non coding RNAs ,circular RNA, splicing, non coding RNAs ,RNA ,Circular ,circular RNA ,Cell Biology ,RNA/genetics ,RNA, Circular ,Molecular Biology ,Biochemistry ,Biotechnology - Abstract
Circular RNAs (circRNAs) are formed in all domains of life and via different mechanisms. There has been an explosion in the number of circRNA papers in recent years; however, as a relatively young field, circRNA biology has an urgent need for common experimental standards for isolating, analyzing, expressing and depleting circRNAs. Here we propose a set of guidelines for circRNA studies based on the authors’ experience. This Perspective will specifically address the major class of circRNAs in Eukarya that are generated by a spliceosome-catalyzed back-splicing event. We hope that the implementation of best practice principles for circRNA research will help move the field forward and allow a better functional understanding of this fascinating group of RNAs.
- Published
- 2022
11. ALS-related FUS mutations alter axon growth in motoneurons and affect HuD/ELAVL4 and FMRP activity
- Author
-
Thomas J. Cunningham, Maria Giovanna Garone, Pietro Fratta, Nicol Birsa, Valeria de Turris, Michela Mochi, Maria Rosito, Remya R. Nair, Mariangela Morlando, Alessandro Rosa, Federico Salaris, and Elizabeth M. C. Fisher
- Subjects
Motor neuron ,QH301-705.5 ,NRN1 ,Mutant ,SOD1 ,Medicine (miscellaneous) ,ELAV-Like Protein 4 ,Biology ,FUS ,iPSC ,motoneuron ,axon ,GAP43 ,HuD ,FMR1 ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Fragile X Mental Retardation Protein ,Mice ,Downregulation and upregulation ,medicine ,Animals ,Humans ,Biology (General) ,Amyotrophic lateral sclerosis ,Gap-43 protein ,Induced pluripotent stem cell ,RNA metabolism ,Motor Neurons ,Messenger RNA ,medicine.disease ,Phenotype ,Axons ,Cell biology ,Induced pluripotent stem cells ,Mechanisms of disease ,Mutation ,biology.protein ,RNA-Binding Protein FUS ,General Agricultural and Biological Sciences - Abstract
Mutations in the RNA-binding protein (RBP) FUS have been genetically associated with the motoneuron disease amyotrophic lateral sclerosis (ALS). Using both human induced pluripotent stem cells and mouse models, we found that FUS-ALS causative mutations affect the activity of two relevant RBPs with important roles in neuronal RNA metabolism: HuD/ELAVL4 and FMRP. Mechanistically, mutant FUS leads to upregulation of HuD protein levels through competition with FMRP for HuD mRNA 3’UTR binding. In turn, increased HuD levels overly stabilize the transcript levels of its targets, NRN1 and GAP43. As a consequence, mutant FUS motoneurons show increased axon branching and growth upon injury, which could be rescued by dampening NRN1 levels. Since similar phenotypes have been previously described in SOD1 and TDP-43 mutant models, increased axonal growth and branching might represent broad early events in the pathogenesis of ALS., Maria Giovanna Garone et al. use iPSC and mouse models to evaluate a mechanistic link between aberrant axonal phenotypes in ALS and the alteration of a cross-regulatory circuitry involving three RNA binding proteins: FUS, HuD and FMRP. Their results suggest NRN1 as a potential therapeutic target for ALS and provide further insight into the pathogenesis of this critical disorder.
- Published
- 2021
12. Study of Circular RNA Expression by Nonradioactive Northern Blot Procedure
- Author
-
Eleonora, D'Ambra and Mariangela, Morlando
- Subjects
RNA, Untranslated ,Gene Expression ,Humans ,RNA Probes ,RNA, Circular ,Blotting, Northern - Abstract
Circular RNAs (circRNAs) are covalently closed transcripts generated by back-splicing reaction. The lack of free ends endows these RNA molecules with high stability thus allowing them to accumulate in tissues and body fluids. They are widely expressed in most organisms, are modulated during development and display tissue-specific expression, resulting particularly enriched in the nervous system. Deregulation of circRNA expression has also been associated with several pathological conditions including neurological diseases and cancer.Here we present a Northern blot procedure that allows the analysis of the expression of bona fide circRNAs through the use of a digoxigenin-labeled RNA probe and the immunodetection of the signals.
- Published
- 2021
13. A longitudinal study defined circulating microRNAs as reliable biomarkers for disease prognosis and progression in ALS human patients
- Author
-
Mario Sabatelli, Antonio Petrucci, Alessio Colantoni, Maurizio Inghilleri, Adriano Chiò, Irene Bozzoni, Gabriella Dobrowolny, Julie Martone, Antonio Musarò, Andrea Calvo, Giulia Bisogni, Irene Casola, Elisa Lepore, Mariangela Morlando, and Bianca Maria Scicchitano
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Longitudinal study ,Immunology ,Disease ,lcsh:RC254-282 ,Article ,Prognostic markers ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Internal medicine ,microRNA ,medicine ,Paralysis ,lcsh:QH573-671 ,Stage (cooking) ,Amyotrophic lateral sclerosis ,lcsh:Cytology ,business.industry ,biomarkers ,Cell Biology ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,ALS, circulating microRNAs, biomarkers ,Muscle atrophy ,Circulating MicroRNA ,030104 developmental biology ,miRNAs ,circulating microRNAs ,Settore BIO/17 - ISTOLOGIA ,medicine.symptom ,ALS ,business ,030217 neurology & neurosurgery - Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease associated with motor neuron degeneration, muscle atrophy and paralysis. To date, multiple panels of biomarkers have been described in ALS patients and murine models. Nevertheless, none of them has sufficient specificity and thus the molecular signature for ALS prognosis and progression remains to be elucidated. Here we overcome this limitation through a longitudinal study, analyzing serum levels of circulating miRNAs, stable molecules that are recently used as promising biomarkers for many types of human disorders, in ALS patients during the progression of the pathology. We performed next-generation sequencing (NGS) analysis and absolute RT quantification of serum samples of ALS patients and healthy controls. The expression levels of five selected miRNAs were quantitatively analyzed during disease progression in each patient and we demonstrated that high levels of miR-206, miR-133a and miR-151a-5p can predict a slower clinical decline of patient functionality. In particular, we found that miR-206 and miR-151a-5p serum levels were significantly up-regulated at the mild stage of ALS pathology, to decrease in the following moderate and severe stages, whereas the expression levels of miR-133a and miR-199a-5p remained low throughout the course of the disease, showing a diagnostic significance in moderate and severe stages for miR-133a and in mild and terminal ones for miR-199a-5p. Moreover, we found that miR-423–3p and 151a-5p were significantly downregulated respectively in mild and terminal stages of the disease. These data suggest that these miRNAs represent potential prognostic markers for ALS disease.
- Published
- 2021
14. Study of Circular RNA Expression by Nonradioactive Northern Blot Procedure
- Author
-
Eleonora D’Ambra and Mariangela Morlando
- Subjects
Immunodetection ,0301 basic medicine ,Nervous system ,Chemistry ,RNA probe ,RNA ,Non-coding RNA ,RNA electrophoresis ,Noncoding RNA ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Circular RNA ,030220 oncology & carcinogenesis ,medicine ,Digoxigenin ,circRNA ,Northern blot ,Northern Blot - Abstract
Circular RNAs (circRNAs) are covalently closed transcripts generated by back-splicing reaction. The lack of free ends endows these RNA molecules with high stability thus allowing them to accumulate in tissues and body fluids. They are widely expressed in most organisms, are modulated during development and display tissue-specific expression, resulting particularly enriched in the nervous system. Deregulation of circRNA expression has also been associated with several pathological conditions including neurological diseases and cancer.Here we present a Northern blot procedure that allows the analysis of the expression of bona fide circRNAs through the use of a digoxigenin-labeled RNA probe and the immunodetection of the signals.
- Published
- 2021
15. Circ-Hdgfrp3 shuttles along neurites and is trapped in aggregates formed by ALS-associated mutant FUS
- Author
-
Irene Bozzoni, Erika Vitiello, Eleonora D’Ambra, Mariangela Morlando, Valentina Silenzi, Sara D’Uva, and Tiziana Santini
- Subjects
0303 health sciences ,Multidisciplinary ,Neurite ,Chemistry ,Science ,Mutant ,Wild type ,RNA ,biological sciences ,cellular neuroscience ,molecular neuroscience ,medicine.disease_cause ,Molecular neuroscience ,Article ,Cellular neuroscience ,Cell biology ,03 medical and health sciences ,Biological sciences ,0302 clinical medicine ,Stress granule ,Cytoplasm ,Circular RNA ,medicine ,030217 neurology & neurosurgery ,Oxidative stress ,030304 developmental biology - Abstract
Summary: CircRNAs belong to a family of RNA molecules which are conserved in evolution, have tissue-specific expression, and are abundant in neuronal cells. Here, we define several features of circ-Hdgfrp3 and describe interesting alterations occurring in motor neurons (MNs) carrying ALS-associated FUS mutations. Through a highly sensitive in situ approach we describe that circ-Hdgfrp3 traffics along neurites, while upon oxidative stress it is retained in the perinuclear region. While in wild-type stressed MNs, circ-Hdgfrp3 localizes in stress granules (SGs), in MNs carrying mutant FUS, a higher proportion of circ-Hdgfrp3 was trapped into cytoplasmic aggregates. Upon stress removal, circ-Hdgfrp3 was easily freed from SGs whereas it was less efficiently released from FUS-aggregates. We found that the human circ-Hdgfrp3 counterpart was also similarly associated to mutant FUS-aggregates in stressed neuronal cells. Overall, the alteration of circ-Hdgfrp3 trafficking adds a further layer of complexity to the role of FUS-aggregates in ALS disease.
- Published
- 2021
16. Emerging Role for Linear and Circular Spermine Oxidase RNAs in Skeletal Muscle Physiopathology
- Author
-
Giulia Baroli, Roberta Ceci, Cristiana Valle, Irene Bozzoni, Jonathan Fernando Reinoso-Sánchez, Paolo Mariottini, Silvia Scaricamazza, Robert Anthony Casero Casero, Mariangela Morlando, Manuela Cervelli, Stefania Sabatini, Guglielmo Duranti, Reinoso-Sanchez, J. F., Baroli, G., Duranti, G., Scaricamazza, S., Sabatini, S., Valle, C., Morlando, M., Casero, R. A., Bozzoni, I., Mariottini, P., Ceci, R., and Cervelli, M.
- Subjects
Male ,RNA, Untranslated ,Muscle Fibers, Skeletal ,lcsh:Chemistry ,Mice ,Superoxide Dismutase-1 ,circRNA ,Amyotrophic lateral sclerosis ,lcsh:QH301-705.5 ,Spectroscopy ,Cells, Cultured ,Oxidoreductases Acting on CH-NH Group Donors ,Myogenesis ,Cell Differentiation ,General Medicine ,Muscle atrophy ,Computer Science Applications ,Cell biology ,CircRNA ,Muscular Atrophy ,medicine.anatomical_structure ,Spermine oxidase ,Female ,medicine.symptom ,C2C12 ,Mice, Transgenic ,Biology ,Catalysis ,Article ,Inorganic Chemistry ,Atrophy ,skeletal muscle atrophy ,amyotrophic lateral sclerosis murine models ,medicine ,Animals ,RNA, Messenger ,Physical and Theoretical Chemistry ,spermine oxidase ,Muscle, Skeletal ,Molecular Biology ,Amyotrophic lateral sclerosis murine model ,Organic Chemistry ,Amyotrophic Lateral Sclerosis ,Skeletal muscle ,RNA ,RNA, Circular ,medicine.disease ,Mice, Inbred C57BL ,Disease Models, Animal ,lcsh:Biology (General) ,lcsh:QD1-999 ,RNA-Binding Protein FUS ,Skeletal muscle atrophy - Abstract
Skeletal muscle atrophy is a pathological condition so far without eective treatment and poorly understood at a molecular level. Emerging evidence suggest a key role for circular RNAs (circRNA) during myogenesis and their deregulation has been reported to be associated with muscle diseases. Spermine oxidase (SMOX), a polyamine catabolic enzyme plays a critical role in muscle dierentiation and the existence of a circRNA arising from SMOX gene has been recently identified. In this study, we evaluated the expression profile of circular and linear SMOX in both C2C12 dierentiation and dexamethasone-induced myotubes atrophy. To validate our findings in vivo their expression levels were also tested in two murine models of amyotrophic lateral sclerosis: SOD1G93A and hFUS+/+, characterized by progressive muscle atrophy. During C2C12 dierentiation, linear and circular SMOX show the same trend of expression. Interestingly, in atrophy circSMOX levels significantly increased compared to the physiological state, in both in vitro and in vivo models. Our study demonstrates that SMOX represents a new player in muscle physiopathology and provides a scientific basis for further investigation on circSMOX RNA as a possible new therapeutic target for the treatment of muscle atrophy.
- Published
- 2020
- Full Text
- View/download PDF
17. ALS-FUS mutation affects the activities of HuD/ELAVL4 and FMRP leading to axon phenotypes in motoneurons
- Author
-
Valeria de Turris, Pietro Fratta, Thomas J. Cunningham, Nicol Birsa, Alessandro Rosa, Maria Rosito, Federico Salaris, Elizabeth M. C. Fisher, Maria Giovanna Garone, Remya R. Nair, Michela Mochi, and Mariangela Morlando
- Subjects
Mutation ,biology ,Mutant ,SOD1 ,medicine.disease_cause ,medicine.disease ,Phenotype ,Cell biology ,medicine.anatomical_structure ,Downregulation and upregulation ,medicine ,biology.protein ,Gap-43 protein ,Amyotrophic lateral sclerosis ,Axon - Abstract
Mutations in the RNA-binding protein (RBPs) FUS have been genetically associated with the motoneuron disease amyotrophic lateral sclerosis (ALS). Using both human induced pluripotent stem cells and mouse models, we found that FUS-ALS causative mutations affect the activity of two relevant RBPs with important roles in neuronal RNA metabolism: HuD/ELAVL4 and FMRP. Mechanistically, mutant FUS leads to upregulation of HuD protein levels through competition with FMRP for HuD mRNA 3’UTR binding. In turn, increased HuD levels overly stabilize the transcript levels of its targets, NRN1 and GAP43. As a consequence, mutant FUS motoneurons show increased axon branching and growth upon injury, which could be rescued by dampening NRN1 levels. Since similar phenotypes have been previously described in SOD1 and TDP-43 mutant models, increased axonal growth and branching might represent broad early events in the pathogenesis of ALS.
- Published
- 2020
18. Modulation of circRNA Metabolism by m6A Modification
- Author
-
Alessio Colantoni, Gaia Di Timoteo, Dario Dattilo, Alessandro Fatica, Irene Bozzoni, Francesca Rossi, Salvatore Oliviero, Danny Incarnato, Alvaro Centrón-Broco, Marco Guarnacci, Mariangela Morlando, and Molecular Genetics
- Subjects
0301 basic medicine ,BIOGENESIS ,cap-independent translation ,M6A ,General Biochemistry, Genetics and Molecular Biology ,A modification ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Circular RNA ,YTHDC1 ,circRNA ,epitrascriptome ,splice ,CIRCULAR RNA ,Messenger RNA ,Chemistry ,circRNA biogenesis ,Translation (biology) ,Cell biology ,back-splicing ,030104 developmental biology ,circRNA translation ,RNA splicing ,METTL3 ,m ,6 ,TRANSLATION ,MESSENGER-RNA ,LEUKEMIA ,030217 neurology & neurosurgery ,Function (biology) ,Biogenesis - Abstract
N6-methyladenosine (m6A) is an RNA modification well-known for its contribution to different processes controlling RNA metabolism, including splicing, stability, and translation of mRNA. Conversely, the role of m6A on the biogenesis and function of circular RNAs (circRNAs) has yet to be addressed. circRNAs belong to a class of covalently closed transcripts produced via a back-splicing reaction whereby a downstream 5' splice donor site fuses to an upstream 3' splice acceptor site. Starting from circ-ZNF609 as a study case, we discover that specific m6As control its accumulation and that METTL3 and YTHDC1 are required to direct the back-splicing reaction. This feature is shared with other circRNAs because we find a significant direct correlation among METTL3 requirement, YTHDC1 binding, and the ability of m6A exons to undergo back-splicing. Finally, because circ-ZNF609 displays the ability to be translated, we show that m6A modifications, through recognition by YTHDF3 and eIF4G2, modulate its translation.
- Published
- 2020
19. SMaRT lncRNA controls translation of a G‐quadruplex‐containing mRNA antagonizing the DHX36 helicase
- Author
-
Julie Martone, Tiziana Santini, Adriano Setti, Mariangela Morlando, Alessio Colantoni, Sama Shamloo, Irene Bozzoni, Alessandro Paiardini, Dacia Dimartino, Davide Mariani, and Francesca Capparelli
- Subjects
lncRNAs ,Biochemistry ,Article ,G-quadruplex ,Myogenesis ,RNA helicases ,RNA–RNA interactions ,DEAD-box RNA Helicases ,Mice ,03 medical and health sciences ,0302 clinical medicine ,DHX36 ,Gene expression ,Genetics ,Animals ,RNA, Messenger ,MLX ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Messenger RNA ,biology ,Chemistry ,DNA Helicases ,Helicase ,RNA ,Translation (biology) ,Articles ,RNA Biology ,RNA Helicase A ,Cell biology ,G-Quadruplexes ,G‐quadruplex ,biology.protein ,RNA, Long Noncoding ,Development & Differentiation ,030217 neurology & neurosurgery - Abstract
Guanine‐quadruplexes (G4) included in RNA molecules exert several functions in controlling gene expression at post‐transcriptional level; however, the molecular mechanisms of G4‐mediated regulation are still poorly understood. Here, we describe a regulatory circuitry operating in the early phases of murine muscle differentiation in which a long non‐coding RNA (SMaRT) base pairs with a G4‐containing mRNA (Mlx‐γ) and represses its translation by counteracting the activity of the DHX36 RNA helicase. The time‐restricted, specific effect of lnc‐SMaRT on the translation of Mlx‐γ isoform modulates the general subcellular localization of total MLX proteins, impacting on their transcriptional output and promoting proper myogenesis and mature myotube formation. Therefore, the circuitry made of lnc‐SMaRT, Mlx‐γ, and DHX36 not only plays an important role in the control of myogenesis but also unravels a molecular mechanism where G4 structures and G4 unwinding activities are regulated in living cells., This study shows that muscle‐specific lnc‐SMaRT regulates early myogenesis. By base‐pairing to the G4 region present in the Mlx‐γ mRNA, lnc‐SMaRT controls its translation in an antagonistic manner to the RNA helicase DHX36.
- Published
- 2020
- Full Text
- View/download PDF
20. Non-coding RNAs in Nervous System Development and Disease
- Author
-
Mariangela Morlando, Beatrice Salvatori, and Silvia Biscarini
- Subjects
0301 basic medicine ,Nervous system ,Cell type ,Review ,Disease ,Computational biology ,Biology ,biomarkers ,circRNAs ,ncRNAs ,neurodegenerative diseases ,neuronal development ,synaptic activity ,therapeutics ,Transcriptome ,Cell and Developmental Biology ,03 medical and health sciences ,0302 clinical medicine ,medicine ,lcsh:QH301-705.5 ,Regulation of gene expression ,Neurodegeneration ,RNA ,Cell Biology ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,030220 oncology & carcinogenesis ,Nucleic acid ,Developmental Biology - Abstract
The rapid advance of RNA sequencing technologies contributed to a deep understanding of transcriptome composition and has allowed the discovery of a large number of non-coding RNAs (ncRNAs). The ability of these RNA molecules to be engaged in intricate and dynamic interactions with proteins and nucleic acids led to a great expansion of gene expression regulation mechanisms. By this matter, ncRNAs contribute to the increase in regulatory complexity that becomes highly specific between tissues and cell types. Among the ncRNAs, long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) are especially abundant in nervous system and have been shown to be implicated in its development, plasticity and aging as well as in neurological disorders. This review provides an overview of how these two diverse classes of ncRNAs control cellular processes during nervous system development, physiology, and disease conditions with particular emphasis on neurodegenerative disorders. The use of ncRNAs as biomarkers, tools, or targets for therapeutic intervention in neurodegeneration are also discussed.
- Published
- 2020
21. The Long Non-coding RNA lnc-31 Interacts with Rock1 mRNA and Mediates Its YB-1-Dependent Translation
- Author
-
Irene Bozzoni, Dacia Dimartino, Monica Ballarino, Julie Martone, Johannes Danner, Gunter Meister, Davide Mariani, Mariangela Morlando, Astrid Bruckmann, and Alessio Colantoni
- Subjects
Genetics and Molecular Biology (all) ,0301 basic medicine ,Myoblast proliferation ,Rock1 ,translation ,Biochemistry ,YB-1 ,lnc-31 ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,03 medical and health sciences ,Mice ,Downregulation and upregulation ,Gene expression ,Protein biosynthesis ,Animals ,RNA, Messenger ,RNA, Small Interfering ,lcsh:QH301-705.5 ,Cell Proliferation ,Messenger RNA ,rho-Associated Kinases ,long non-coding RNA ,Chemistry ,RNA ,Long non-coding RNA ,Cell biology ,030104 developmental biology ,proteasome ,lcsh:Biology (General) ,protein stability ,Protein Biosynthesis ,cell cycle ,myogenesis ,biochemistry ,genetics and Molecular Biology (all) ,Translational Activation ,RNA Interference ,RNA, Long Noncoding ,Biochemistry, Genetics and Molecular Biology (all) ,5' Untranslated Regions ,Protein Binding ,Transcription Factors - Abstract
Summary Cytoplasmic long non-coding RNAs have been shown to act at many different levels to control post-transcriptional gene expression, although their role in translational control is poorly understood. Here, we show that lnc-31, a non-coding RNA required for myoblast proliferation, promotes ROCK1 protein synthesis by stabilizing its translational activator, YB-1. We find that lnc-31 binds to the Rock1 mRNA as well as to the YB-1 protein and that translational activation requires physical interaction between the two RNA species. These results suggest a localized effect of YB-1 stabilization on the Rock1 mRNA. ROCK1 upregulation by lnc-31, in proliferative conditions, correlates well with the differentiation-repressing activity of ROCK1. We also show that, upon induction of differentiation, the downregulation of lnc-31, in conjunction with miR-152 targeting of Rock1, establishes a regulatory loop that reinforces ROCK1 repression and promotes myogenesis., Graphical Abstract, Highlights • lnc-31 sustains myoblast proliferation, counteracting differentiation • lnc-31 binds to Rock1 mRNA and YB-1 protein • Rock-1 translation is favored through its interaction with lnc-31 and YB-1 protein • lnc-31 counteracts YB-1 protein degradation, thus promoting Rock1 translation, Dimartino et al. demonstrate that lnc-31 is required to sustain myoblast proliferation. lnc-31 interacts with Rock1 mRNA, an inhibitor of differentiation, and promotes its translation. This activity is strengthened by binding of the translational regulator YB-1 and its lnc-31-dependent stabilization.
- Published
- 2018
22. FUS affects circular RNA expression in murine embryonic stem cell-derived motor neurons
- Author
-
Elisa Caffarelli, Lei Lu, Lorenzo Errichelli, Stefano Dini Modigliani, Ivano Legnini, Pietro Laneve, Irene Bozzoni, Neil A. Shneider, Rebecca Scarfò, Giovanna Peruzzi, Riccardo De Santis, Alessandro Rosa, Mariangela Morlando, Alessio Colantoni, and Davide Capauto
- Subjects
0301 basic medicine ,Genetics and Molecular Biology (all) ,Cellular differentiation ,RNA-binding protein ,General Physics and Astronomy ,Biochemistry ,Mice ,FUS protein ,RNA interference ,embryonic stem cell-derived motor neurons ,Regulation of gene expression ,Motor Neurons ,Multidisciplinary ,General Commentary ,Chemistry (all) ,Cell Differentiation ,Mouse Embryonic Stem Cells ,Exons ,Spinal Cord ,motor neuron disease ,Sequence Analysis ,circular RNAs ,Protein Binding ,RNA Splicing ,Science ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,splicing ,Physics and Astronomy (all) ,Circular RNA ,Animals ,mouse ,FUS ,Sequence Analysis, RNA ,Intron ,Gene Deletion ,Gene Expression Regulation ,Introns ,Mutation ,RNA ,RNA-Binding Protein FUS ,Biochemistry, Genetics and Molecular Biology (all) ,circular RNA ,General Chemistry ,RNA, Circular ,Embryonic stem cell ,Molecular biology ,030104 developmental biology ,Biogenesis ,Neuroscience - Abstract
The RNA-binding protein FUS participates in several RNA biosynthetic processes and has been linked to the pathogenesis of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia. Here we report that FUS controls back-splicing reactions leading to circular RNA (circRNA) production. We identified circRNAs expressed in in vitro-derived mouse motor neurons (MNs) and determined that the production of a considerable number of these circRNAs is regulated by FUS. Using RNAi and overexpression of wild-type and ALS-associated FUS mutants, we directly correlate the modulation of circRNA biogenesis with alteration of FUS nuclear levels and with putative toxic gain of function activities. We also demonstrate that FUS regulates circRNA biogenesis by binding the introns flanking the back-splicing junctions and that this control can be reproduced with artificial constructs. Most circRNAs are conserved in humans and specific ones are deregulated in human-induced pluripotent stem cell-derived MNs carrying the FUSP525L mutation associated with ALS.
- Published
- 2017
23. Modulation of circRNA Metabolism by m
- Author
-
Gaia, Di Timoteo, Dario, Dattilo, Alvaro, Centrón-Broco, Alessio, Colantoni, Marco, Guarnacci, Francesca, Rossi, Danny, Incarnato, Salvatore, Oliviero, Alessandro, Fatica, Mariangela, Morlando, and Irene, Bozzoni
- Subjects
Male ,Alternative Splicing ,Adenosine ,HEK293 Cells ,Child, Preschool ,Humans ,Female ,Nerve Tissue Proteins ,Methyltransferases ,RNA Splicing Factors ,RNA, Circular ,HeLa Cells - Abstract
N
- Published
- 2019
24. Circ-ZNF609 regulates G1-S progression in rhabdomyosarcoma
- Author
-
Gaia Di Timoteo, Dario Dattilo, Francesca Rossi, Tiziana Santini, Alessio Colantoni, Ivano Legnini, Francesca Megiorni, Irene Bozzoni, Carlo Dominici, and Mariangela Morlando
- Subjects
0301 basic medicine ,Cancer Research ,Cell ,Regulator ,circular rna ,Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Cell Line, Tumor ,medicine ,Genetics ,Humans ,Rhabdomyosarcoma, Embryonal ,Rhabdomyosarcoma ,Gene ,Molecular Biology ,Rhabdomyosarcoma, Alveolar ,Regulation of gene expression ,Cell Cycle ,RNA, Circular ,Cell cycle ,medicine.disease ,rhabdomyosarcoma ,cell proliferation ,g1-s transition ,Up-Regulation ,Cell biology ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Cell culture ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,RNA - Abstract
Circular RNAs (circRNAs) represent a class of covalently closed RNAs, derived from non-canonical splicing events, which are expressed in all eukaryotes and often conserved among different species. We previously showed that the circRNA originating from the ZNF609 locus (circ-ZNF609) acts as a crucial regulator of human primary myoblast growth: indeed, the down-regulation of the circRNA, and not of its linear counterpart, strongly reduced the proliferation rate of in vitro cultured myoblasts. To deepen our knowledge about circ-ZNF609 role in cell cycle regulation, we studied its expression and function in rhabdomyosarcoma (RMS), a pediatric skeletal muscle malignancy. We found that circ-ZNF609 is up-regulated in biopsies from the two major RMS subtypes, embryonal (ERMS) and alveolar (ARMS). Moreover, we discovered that in an ERMS-derived cell line circ-ZNF609 knock-down induced a specific block at the G1-S transition, a strong decrease of p-Akt protein level and an alteration of the pRb/Rb ratio. Regarding p-Akt, we were able to show that circ-ZNF609 acts by counteracting p-Akt proteasome-dependent degradation, thus working as a new regulator of cell proliferation-related pathways. As opposed to ERMS-derived cells, the circRNA depletion had no cell cycle effects in ARMS-derived cells. Since in these cells the p53 gene resulted down-regulated, with a concomitant up-regulation of its cell cycle-related target genes, we suggest that this could account for the lack of circ-ZNF609 effect in ARMS.
- Published
- 2019
25. Increased FUS levels in astrocytes leads to astrocyte and microglia activation and neuronal death
- Author
-
Giuseppe Lupo, Irene Bozzoni, Maria Antonietta Ajmone-Cat, Camilla Toselli, Luisa Minghetti, Stefano Biagioni, Mariangela Morlando, Emanuela Monni, Angela Onori, Eleonora Stronati, Emanuele Cacci, and Zaal Kokaia
- Subjects
0301 basic medicine ,Untranslated region ,Programmed cell death ,Neuroinflammation ,Amyotrophic lateral sclerosis ,Neural stem cells ,Astrocyes ,microglia ,lcsh:Medicine ,Biology ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,Coding region ,lcsh:Science ,Ribonucleoprotein ,Motor Neurons ,Neurons ,Multidisciplinary ,Cell Death ,Microglia ,Gene Expression Profiling ,lcsh:R ,Neurodegeneration ,medicine.disease ,Cell biology ,Protein Transport ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Astrocytes ,Mutation ,RNA-Binding Protein FUS ,lcsh:Q ,Disease Susceptibility ,Inflammation Mediators ,Biomarkers ,030217 neurology & neurosurgery ,Astrocyte - Abstract
Mutations of Fused in sarcoma (FUS), a ribonucleoprotein involved in RNA metabolism, have been found associated with both familial and sporadic cases of amyotrophic lateral sclerosis (ALS). Notably, besides mutations in the coding sequence, also mutations into the 3′ untranslated region, leading to increased levels of the wild-type protein, have been associated with neuronal death and ALS pathology, in ALS models and patients. The mechanistic link between altered FUS levels and ALS-related neurodegeneration is far to be elucidated, as well as the consequences of elevated FUS levels in the modulation of the inflammatory response sustained by glial cells, a well-recognized player in ALS progression. Here, we studied the effect of wild-type FUS overexpression on the responsiveness of mouse and human neural progenitor-derived astrocytes to a pro-inflammatory stimulus (IL1β) used to mimic an inflammatory environment. We found that astrocytes with increased FUS levels were more sensitive to IL1β, as shown by their enhanced expression of inflammatory genes, compared with control astrocytes. Moreover, astrocytes overexpressing FUS promoted neuronal cell death and pro-inflammatory microglia activation. We conclude that overexpression of wild-type FUS intrinsically affects astrocyte reactivity and drives their properties toward pro-inflammatory and neurotoxic functions, suggesting that a non-cell autonomous mechanism can support neurodegeneration in FUS-mutated animals and patients.
- Published
- 2019
26. Non-coding RNAs in muscle differentiation and musculoskeletal disease
- Author
-
Mariangela Morlando, Irene Bozzoni, Monica Ballarino, and Alessandro Fatica
- Subjects
Genetic Markers ,0301 basic medicine ,RNA, Untranslated ,Myoblasts, Skeletal ,Cellular differentiation ,Coding (therapy) ,Review ,Computational biology ,Biology ,Muscle Development ,Models, Biological ,Myoblasts ,Transcriptome ,Mice ,03 medical and health sciences ,Animals ,Biomarkers ,Cell Differentiation ,Humans ,MicroRNAs ,Muscle, Skeletal ,Musculoskeletal Diseases ,RNA, Long Noncoding ,Medicine (all) ,Models ,microRNA ,medicine ,Myocyte ,duchenne muscular-dystrophy ,competing endogenous rna ,skeletal-muscle ,myoblast differentiation ,myogenic differentiation ,deacetylase inhibitors ,human rhabdomyosarcoma ,cell-differentiation ,regulatory networks ,serum biomarkers ,Genetics ,Untranslated ,RNA ,Skeletal muscle ,Skeletal ,General Medicine ,Biological ,030104 developmental biology ,medicine.anatomical_structure ,Muscle ,Long Noncoding ,Identification (biology) - Abstract
RNA is likely to be the most rediscovered macromolecule in biology. Periodically, new non-canonical functions have been ascribed to RNA, such as the ability to act as a catalytic molecule or to work independently from its coding capacity. Recent annotations show that more than half of the transcriptome encodes for RNA molecules lacking coding activity. Here we illustrate how these transcripts affect skeletal muscle differentiation and related disorders. We discuss the most recent scientific discoveries that have led to the identification of the molecular circuitries that are controlled by RNA during the differentiation process and that, when deregulated, lead to pathogenic events. These findings will provide insights that can aid in the development of new therapeutic interventions for muscle diseases.
- Published
- 2016
27. Deficiency in the nuclear long noncoding <scp>RNA</scp> Charme causes myogenic defects and heart remodeling in mice
- Author
-
Andrea Cipriano, Dònal O’ Carroll, Monica Ballarino, Carmine Nicoletti, Claudia Carrieri, Rossella Tita, Antonio Musarò, Alessio Colantoni, Tiziana Santini, Irene Bozzoni, Fabio Desideri, and Mariangela Morlando
- Subjects
0301 basic medicine ,lncRNAs ,CRISPR/Cas9 ,epigenetic control ,heart development ,myogenesis ,Neuroscience (all) ,Molecular Biology ,Biochemistry, Genetics and Molecular Biology (all) ,Immunology and Microbiology (all) ,Biology ,incRNAs ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Downregulation and upregulation ,microRNA ,biochemistry ,Myocyte ,Gene ,neuroscience (all) ,molecular biology ,biochemistry, genetics and molecular biology (all) ,immunology and microbiology (all) ,General Immunology and Microbiology ,Heart development ,Myogenesis ,genetics and molecular biology (all) ,General Neuroscience ,Phenotype ,Long non-coding RNA ,Cell biology ,030104 developmental biology - Abstract
Myogenesis is a highly regulated process that involves the conversion of progenitor cells into multinucleated myofibers. Besides proteins and miRNAs, long noncoding RNAs (lncRNAs) have been shown to participate in myogenic regulatory circuitries. Here, we characterize a murine chromatin‐associated muscle‐specific lncRNA, Charme , which contributes to the robustness of the myogenic program in vitro and in vivo . In myocytes, Charme depletion triggers the disassembly of a specific chromosomal domain and the downregulation of myogenic genes contained therein. Notably, several Charme ‐sensitive genes are associated with human cardiomyopathies and Charme depletion in mice results in a peculiar cardiac remodeling phenotype with changes in size, structure, and shape of the heart. Moreover, the existence of an orthologous transcript in human, regulating the same subset of target genes, suggests an important and evolutionarily conserved function for Charme . Altogether, these data describe a new example of a chromatin‐associated lncRNA regulating the robustness of skeletal and cardiac myogenesis.
- Published
- 2018
28. Correction for Ballarino et al., 'Novel Long Noncoding RNAs (lncRNAs) in Myogenesis: a miR-31 Overlapping lncRNA Transcript Controls Myoblast Differentiation'
- Author
-
Tiziana Santini, Andrea Cipriano, Bisceglie L, Daniel D'Andrea, Irene Bozzoni, Luigi Grassi, Cazzella, Mariangela Morlando, Chiara Pinnarò, Anna Tramontano, and Monica Ballarino
- Subjects
Mice, Transgenic ,Biology ,Muscle Development ,Myoblasts ,Mice ,RNA Precursors ,Animals ,Humans ,Myocyte ,Author Correction ,Molecular Biology ,Cell Proliferation ,Myogenesis ,Gene Expression Profiling ,Cell Differentiation ,Muscle, Smooth ,Articles ,Cell Biology ,Biological Evolution ,Muscle, Striated ,Cell biology ,Muscular Dystrophy, Duchenne ,mir-31 ,MicroRNAs ,Gene Expression Regulation ,RNA, Long Noncoding ,Transcriptome - Abstract
Transcriptome analysis allowed the identification of new long noncoding RNAs differentially expressed during murine myoblast differentiation. These transcripts were classified on the basis of their expression under proliferating versus differentiated conditions, muscle-restricted activation, and subcellular localization. Several species displayed preferential expression in dystrophic (mdx) versus wild-type muscles, indicating their possible link with regenerative processes. One of the identified transcripts, lnc-31, even if originating from the same nuclear precursor of miR-31, is produced by a pathway mutually exclusive. We show that lnc-31 and its human homologue hsa-lnc-31 are expressed in proliferating myoblasts, where they counteract differentiation. In line with this, both species are more abundant in mdx muscles and in human Duchenne muscular dystrophy (DMD) myoblasts, than in their normal counterparts. Altogether, these data suggest a crucial role for lnc-31 in controlling the differentiation commitment of precursor myoblasts and indicate that its function is maintained in evolution despite the poor sequence conservation with the human counterpart.
- Published
- 2018
29. Deficiency in the nuclear long noncoding RNA
- Author
-
Monica, Ballarino, Andrea, Cipriano, Rossella, Tita, Tiziana, Santini, Fabio, Desideri, Mariangela, Morlando, Alessio, Colantoni, Claudia, Carrieri, Carmine, Nicoletti, Antonio, Musarò, Dònal O', Carroll, and Irene, Bozzoni
- Subjects
Vascular Biology & Angiogenesis ,Ventricular Remodeling ,Muscle Fibers, Skeletal ,lncRNAs ,Articles ,heart development ,Muscle Development ,epigenetic control ,Chromatin, Epigenetics, Genomics & Functional Genomics ,RNA Biology ,Article ,Mice ,Animals ,Humans ,Myocytes, Cardiac ,RNA, Long Noncoding ,myogenesis ,CRISPR/Cas9 - Abstract
Myogenesis is a highly regulated process that involves the conversion of progenitor cells into multinucleated myofibers. Besides proteins and miRNAs, long noncoding RNAs (lncRNAs) have been shown to participate in myogenic regulatory circuitries. Here, we characterize a murine chromatin‐associated muscle‐specific lncRNA, Charme, which contributes to the robustness of the myogenic program in vitro and in vivo. In myocytes, Charme depletion triggers the disassembly of a specific chromosomal domain and the downregulation of myogenic genes contained therein. Notably, several Charme‐sensitive genes are associated with human cardiomyopathies and Charme depletion in mice results in a peculiar cardiac remodeling phenotype with changes in size, structure, and shape of the heart. Moreover, the existence of an orthologous transcript in human, regulating the same subset of target genes, suggests an important and evolutionarily conserved function for Charme. Altogether, these data describe a new example of a chromatin‐associated lncRNA regulating the robustness of skeletal and cardiac myogenesis.
- Published
- 2018
30. A Regulatory Circuitry Between Gria2, miR-409, and miR-495 Is Affected by ALS FUS Mutation in ESC-Derived Motor Neurons
- Author
-
Pietro Laneve, Irene Bozzoni, Elisa Caffarelli, Neil A. Shneider, Alessio Colantoni, Davide Capauto, Lei Lu, Silvia Biscarini, Mariangela Morlando, Tiziana Santini, and Giovanna Peruzzi
- Subjects
0301 basic medicine ,Messenger ,RNA-binding protein ,Gria2 ,medicine.disease_cause ,ALS ,Excitotoxicity ,FUS ,MicroRNAs ,Motor neurons ,RNA binding proteins ,Cellular and Molecular Neuroscience ,Mice ,Models ,Receptors ,AMPA ,GRIA2 ,Amyotrophic lateral sclerosis ,Regulation of gene expression ,Mutation ,biology ,Cell Differentiation ,Mouse Embryonic Stem Cells ,differentiation ,Cell biology ,Spinal Cord ,Neurology ,Neuroscience (miscellaneous) ,Down-Regulation ,Models, Biological ,Article ,03 medical and health sciences ,Downregulation and upregulation ,microRNA ,medicine ,Amyotrophic Lateral Sclerosis ,Animals ,Gene Expression Profiling ,Gene Expression Regulation ,Motor Neurons ,Protein Subunits ,RNA, Messenger ,RNA-Binding Protein FUS ,Receptors, AMPA ,medicine.disease ,Biological ,030104 developmental biology ,biology.protein ,RNA ,transcriptome - Abstract
Mutations in fused in sarcoma (FUS) cause amyotrophic lateral sclerosis (ALS). FUS is a multifunctional protein involved in the biogenesis and activity of several types of RNAs, and its role in the pathogenesis of ALS may involve both direct effects of disease-associated mutations through gain- and loss-of-function mechanisms and indirect effects due to the cross talk between different classes of FUS-dependent RNAs. To explore how FUS mutations impinge on motor neuron-specific RNA-based circuitries, we performed transcriptome profiling of small and long RNAs of motor neurons (MNs) derived from mouse embryonic stem cells carrying a FUS-P517L knock-in mutation, which is equivalent to human FUS-P525L, associated with a severe and juvenile-onset form of ALS. Combining ontological, predictive and molecular analyses, we found an inverse correlation between several classes of deregulated miRNAs and their corresponding mRNA targets in both homozygous and heterozygous P517L MNs. We validated a circuitry in which the upregulation of miR-409-3p and miR-495-3p, belonging to a brain-specific miRNA subcluster implicated in several neurodevelopmental disorders, produced the downregulation of Gria2, a subunit of the glutamate α‐amino‐3‐hydroxy‐5‐methyl-4-isoxazole propionic acid (AMPA) receptor with a significant role in excitatory neurotransmission. Moreover, we found that FUS was involved in mediating such miRNA repression. Gria2 alteration has been proposed to be implicated in MN degeneration, through disturbance of Ca2+ homeostasis, which triggers a cascade of damaging “excitotoxic” events. The molecular cross talk identified highlights a role for FUS in excitotoxicity and in miRNA-dependent regulation of Gria2. This circuitry also proved to be deregulated in heterozygosity, which matches the human condition perfectly. Electronic supplementary material The online version of this article (10.1007/s12035-018-0884-4) contains supplementary material, which is available to authorized users.
- Published
- 2018
31. Alteration of epigenetic regulation by long noncoding RNAs in cancer
- Author
-
Alessandro Fatica and Mariangela Morlando
- Subjects
0301 basic medicine ,Review ,Biology ,medicine.disease_cause ,Catalysis ,Epigenesis, Genetic ,lcsh:Chemistry ,Inorganic Chemistry ,03 medical and health sciences ,Genetic ,medicine ,Histone code ,Animals ,Humans ,Epigenetics ,Physical and Theoretical Chemistry ,lcsh:QH301-705.5 ,Molecular Biology ,Spectroscopy ,Cancer ,Regulation of gene expression ,Neoplastic ,Organic Chemistry ,Computer Science Applications1707 Computer Vision and Pattern Recognition ,General Medicine ,medicine.disease ,Chromatin ,LncRNA ,Computer Science Applications ,Cell biology ,Gene Expression Regulation, Neoplastic ,cancer ,chromatin ,epigenetics ,lncRNA ,Histone Code ,RNA, Long Noncoding ,030104 developmental biology ,lcsh:Biology (General) ,lcsh:QD1-999 ,Gene Expression Regulation ,RNA ,Human genome ,Long Noncoding ,Carcinogenesis ,Function (biology) ,Epigenesis - Abstract
Long noncoding RNAs (lncRNAs) are important regulators of the epigenetic status of the human genome. Besides their participation to normal physiology, lncRNA expression and function have been already associated to many diseases, including cancer. By interacting with epigenetic regulators and by controlling chromatin topology, their misregulation may result in an aberrant regulation of gene expression that may contribute to tumorigenesis. Here, we review the functional role and mechanisms of action of lncRNAs implicated in the aberrant epigenetic regulation that has characterized cancer development and progression.
- Published
- 2018
32. A Feedforward Regulatory Loop between HuR and the Long Noncoding RNA linc-MD1 Controls Early Phases of Myogenesis
- Author
-
Ivano Legnini, Irene Bozzoni, Mariangela Morlando, Alessandro Fatica, Arianna Mangiavacchi, Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia (IIT), Department of Biology and Biotechnology 'Charles Darwin', Institut Pasteur, Fondation Cenci Bolognetti - Istituto Pasteur Italia, Fondazione Cenci Bolognetti, Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP)-Università degli Studi di Roma 'La Sapienza' = Sapienza University [Rome], Réseau International des Instituts Pasteur (RIIP), and This work was partially supported by grants from Telethon (GGP11149), Epigen, Parent Project Italia, AIRC, IIT 'SEED,' FIRB, and PRIN.
- Subjects
MESH: Cell Differentiation ,Cytoplasm ,MESH: Feedback, Physiological ,Physiological ,Cellular differentiation ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Biology ,Muscle Development ,Feedback ,hur ,long non coding rna ,micro rna ,Cell Line ,Mice ,Short Article ,Animals ,Cell Differentiation ,ELAV Proteins ,Feedback, Physiological ,MicroRNAs ,RNA, Long Noncoding ,microRNA ,MESH: Animals ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,MESH: Mice ,[SDV.BDD]Life Sciences [q-bio]/Development Biology ,Molecular Biology ,Drosha ,Genetics ,Myogenesis ,MESH: Cytoplasm ,MESH: Hu Paraneoplastic Encephalomyelitis Antigens ,RNA ,Cell Biology ,Long non-coding RNA ,MESH: Cell Line ,MESH: RNA, Long Noncoding ,MESH: Muscle Development ,Long Noncoding ,MESH: MicroRNAs ,Biogenesis - Abstract
Summary The muscle-specific long noncoding RNA linc-MD1 was shown to be expressed during early phases of muscle differentiation and to trigger the switch to later stages by acting as a sponge for miR-133 and miR-135. Notably, linc-MD1 is also the host transcript of miR-133b, and their biogenesis is mutually exclusive. Here, we describe that this alternative synthesis is controlled by the HuR protein, which favors linc-MD1 accumulation through its ability to bind linc-MD1 and repress Drosha cleavage. We show that HuR is under the repressive control of miR-133 and that the sponging activity of linc-MD1 consolidates HuR expression in a feedforward positive loop. Finally, we show that HuR also acts in the cytoplasm, reinforcing linc-MD1 sponge activity by cooperating for miRNA recruitment. An increase in miR-133 synthesis, mainly from the two unrelated miR-133a coding genomic loci, is likely to trigger the exit from this circuitry and progression to later differentiation stages., Graphical Abstract, Highlights • A feedforward positive loop exists between linc-MD1 and HuR during myogenesis • HuR controls the relative biogenesis of miR-133b and its host linc-MD1 RNA • Linc-MD1, by sponging miR-133, alleviates its repression on HuR expression • Cytoplasmic HuR reinforces linc-MD1 activity by cooperating for miRNA recruitment, linc-MD1 and miR-133 are alternatively processed from the same precursor RNA. These RNAs play opposing roles in early phases of myogenesis. Legnini et al. now show that the balance between the RNAs is regulated by HuR, which inhibits generation of miR-133 by inhibiting microprocessor activity on the precursor RNA.
- Published
- 2014
33. Exploring the Regulatory Role of Circular RNAs in Neurodegenerative Disorders
- Author
-
Davide Capauto, Mariangela Morlando, and Eleonora D’Ambra
- Subjects
Nervous system ,Review ,Biology ,Catalysis ,lcsh:Chemistry ,Inorganic Chemistry ,neurodegenerative disease ,Gene expression ,medicine ,Animals ,Humans ,Parkinson ,RNA Processing, Post-Transcriptional ,Physical and Theoretical Chemistry ,lcsh:QH301-705.5 ,Molecular Biology ,Spectroscopy ,nervous system ,Organic Chemistry ,Disease progression ,Neurodegenerative Diseases ,RNA, Circular ,General Medicine ,Neuronal pathway ,Computer Science Applications ,medicine.anatomical_structure ,lcsh:Biology (General) ,lcsh:QD1-999 ,Dynamic modulation ,Synaptic plasticity ,Alzheimer ,circRNAs ,ALS ,Neuroscience ,Biomarkers ,Biogenesis ,Function (biology) - Abstract
Circular RNAs (circRNAs) are a distinctive class of regulatory non-coding RNAs characterised by the presence of covalently closed ends. They are evolutionary conserved molecules, and although detected in different tissues, circRNAs resulted specifically enriched in the nervous system. Recent studies have shown that circRNAs are dynamically modulated during neuronal development and aging, that circRNAs are enriched at synaptic levels and resulted modulated after synaptic plasticity induction. This has suggested that circRNAs might play an important role in neuronal specification and activity. Despite the exact function of circRNAs is still poorly understood, emerging evidence indicates that circRNAs have important regulatory functions that might extensively contribute to the dynamic modulation of gene expression that supports neuronal pathways. More interestingly, deregulation of circRNAs expression has been linked with various pathological conditions. In this review, we describe current advances in the field of circRNA biogenesis and function in the nervous system both in physiological and in pathological conditions, and we specifically lay out their association with neurodegenerative diseases. Furthermore, we discuss the opportunity to exploit circRNAs for innovative therapeutic approaches and, due to their high stability, to use circRNAs as suitable biomarkers for diagnosis and disease progression.
- Published
- 2019
34. Non Coding RNA in Muscle Differentiation and Disease
- Author
-
Alessandro Rosa, Mariangela Morlando, Irene Bozzoni, Alessandro Fatica, and Elisa Caffarelli
- Subjects
Cellular differentiation ,muscle differentiation ,Biology ,Genome ,noncoding RNA ,microRNA ,Humans ,Gene Expression Regulation ,MicroRNAs ,Muscle, Skeletal ,RNA, Long Noncoding ,Cell Differentiation ,Orthopedics and Sports Medicine ,Regulation of gene expression ,Genetics ,Messenger RNA ,RNA ,Skeletal ,General Medicine ,Non-coding RNA ,Long non-coding RNA ,Emergency Medicine ,Muscle ,Long Noncoding - Abstract
Non coding RNAs have provided in the last decades a very exciting research field with the discovery that a largely unexplored fraction of our genome encodes for RNA without protein coding activity. Here we revise the current knowledge of how non coding RNAs impact on muscle differentiation and homeostasis in normal and disease conditions and how they can provide powerful tools for therapeutic interventions and disease diagnosis. Moreover, we discuss new insights into additional mechanisms of post-transcriptional regulation involving a new class of long non coding RNAs shown to impact on the distribution of microRNA molecules on their mRNA targets.
- Published
- 2013
35. FUS stimulates microRNA biogenesis by facilitating co-transcriptional Drosha recruitment
- Author
-
Alessandro Rosa, Elisa Caffarelli, Mariangela Morlando, Valerio Di Carlo, Giulia Torrelli, Irene Bozzoni, and Stefano Dini Modigliani
- Subjects
0303 health sciences ,General Immunology and Microbiology ,Immunoprecipitation ,General Neuroscience ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Chromatin ,03 medical and health sciences ,0302 clinical medicine ,microRNA ,Cancer research ,biology.protein ,Ribonuclease III ,Molecular Biology ,Chromatin immunoprecipitation ,030217 neurology & neurosurgery ,Drosha ,Biogenesis ,030304 developmental biology ,RNA-Binding Protein FUS - Abstract
microRNA abundance has been shown to depend on the amount of the microprocessor components or, in some cases, on specific auxiliary co-factors. In this paper, we show that the FUS/TLS (fused in sarcoma/translocated in liposarcoma) protein, associated with familial forms of Amyotrophic Lateral Sclerosis (ALS), contributes to the biogenesis of a specific subset of microRNAs. Among them, species with roles in neuronal function, differentiation and synaptogenesis were identified. We also show that FUS/TLS is recruited to chromatin at sites of their transcription and binds the corresponding pri-microRNAs. Moreover, FUS/TLS depletion leads to decreased Drosha level at the same chromatin loci. Limited FUS/TLS depletion leads to a reduced microRNA biogenesis and we suggest a possible link between FUS mutations affecting nuclear/cytoplasmic partitioning of the protein and altered neuronal microRNA biogenesis in ALS pathogenesis.
- Published
- 2012
36. Coupled RNA processing and transcription of intergenic primary microRNAs
- Author
-
Marcella Marchioni, Francesca Pagano, Mariangela Morlando, Irene Bozzoni, Erika Girardi, Davide Cacchiarelli, Nick J. Proudfoot, Monica Ballarino, Ballarino, Monica, Pagano, Francesca, Girardi, Erika, Morlando, Mariangela, Cacchiarelli, Davide, Marchioni, Marcella, Proudfoot, Nicholas J., and Bozzoni, Irene
- Subjects
Ribonuclease III ,RNA Processing ,DGCR8 ,Messenger ,Post-Transcriptional ,RNA polymerase II ,RNA-Binding Protein ,Primary transcript ,HeLa Cell ,Cell Line ,Microprocessor complex ,Promoter Regions ,Genetic ,Small Nuclear ,Cell Line, Tumor ,RNA, Small Nuclear ,Humans ,RNA, Messenger ,RNA Processing, Post-Transcriptional ,Promoter Regions, Genetic ,RNA polymerase II holoenzyme ,Molecular Biology ,Drosha ,Tumor ,biology ,General transcription factor ,Protein ,Proteins ,RNA-Binding Proteins ,MicroRNA ,Cell Biology ,Articles ,Non-coding RNA ,Molecular biology ,Exoribonuclease ,MicroRNAs ,Exoribonucleases ,HeLa Cells ,RNA Polymerase II ,biology.protein ,RNA ,Human - Abstract
The first step in microRNA (miRNA) biogenesis occurs in the nucleus and is mediated by the Microprocessor complex containing the RNase III-like enzyme Drosha and its cofactor DGCR8. Here we show that the 5'-->3' exonuclease Xrn2 associates with independently transcribed miRNAs and, in combination with Drosha processing, attenuates transcription in downstream regions. We suggest that, after Drosha cleavage, a torpedo-like mechanism acts on nascent long precursor miRNAs, whereby Xrn2 exonuclease degrades the RNA polymerase II-associated transcripts inducing its release from the template. While involved in primary transcript termination, this attenuation effect does not restrict clustered miRNA expression, which, in the majority of cases, is separated by short spacers. We also show that transcripts originating from a miRNA promoter are retained on the chromatin template and are more efficiently processed than those produced from mRNA or snRNA Pol II-dependent promoters. These data imply that coupling between transcription and processing promotes efficient expression of independently transcribed miRNAs.
- Published
- 2016
37. The lack of the Celf2a splicing factor converts a Duchenne genotype into a Becker phenotype
- Author
-
E. Picillo, Olga Sthandier, Ivano Legnini, Irene Bozzoni, Francesca Briganti, Julie Martone, Mariangela Morlando, L. Politano, Martone, J., Briganti, F., Legnini, I., Morlando, M., Picillo, E., Sthandier, O., Politano, Luisa, and Bozzoni, I.
- Subjects
0301 basic medicine ,Male ,Genetics and Molecular Biology (all) ,Duchenne muscular dystrophy ,General Physics and Astronomy ,Biochemistry ,Dystrophin ,Exon ,Genotype-phenotype distinction ,CELF Proteins ,Celf2a ,Muscular Dystrophy ,Muscular dystrophy ,Genetics ,Multidisciplinary ,biology ,Chemistry (all) ,Exons ,Duchenne Muscular Distrophy ,Phenotype ,3. Good health ,Adolescent ,Genotype ,Humans ,Muscular Dystrophy, Duchenne ,Nerve Tissue Proteins ,RNA Splicing ,Biochemistry, Genetics and Molecular Biology (all) ,Physics and Astronomy (all) ,RNA splicing ,splicing ,musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,Science ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Splicing factor ,medicine ,General Chemistry ,medicine.disease ,Duchenne ,Molecular biology ,nervous system diseases ,030104 developmental biology ,biology.protein - Abstract
Substitutions, deletions and duplications in the dystrophin gene lead to either the severe Duchenne muscular dystrophy (DMD) or mild Becker muscular dystrophy depending on whether out-of-frame or in-frame transcripts are produced. We identified a DMD case (GSΔ44) where the correlation between genotype and phenotype is not respected, even if carrying a typical Duchenne mutation (exon 44 deletion) a Becker-like phenotype was observed. Here we report that in this patient, partial restoration of an in-frame transcript occurs by natural skipping of exon 45 and that this is due to the lack of Celf2a, a splicing factor that interacts with exon 45 in the dystrophin pre-mRNA. Several experiments are presented that demonstrate the central role of Celf2a in controlling exon 45 splicing; our data point to this factor as a potential target for the improvement of those DMD therapeutic treatments, which requires exon 45 skipping., Muscular Dystrophy can be caused by mutations in the dystrophin gene, causing the severe Duchenne form or the mild Becker form depending on if the transcript is in or out-of-frame. Here the authors identify a Duchenne-type mutation that gives a Becker-like phenotype due to skipping of exon 45.
- Published
- 2016
38. ALS mutant FUS proteins are recruited into stress granules in induced pluripotent stem cell-derived motoneurons
- Author
-
Mariangela Morlando, Riccardo De Santis, Irene Bozzoni, Jessica Lenzi, Virginia Caliendo, Valeria de Turris, Andrea Calvo, Pietro Laneve, Alessandro Rosa, and Adriano Chiò
- Subjects
Genetics and Molecular Biology (all) ,Cellular differentiation ,Mutant ,lcsh:Medicine ,Medicine (miscellaneous) ,medicine.disease_cause ,Biochemistry ,0302 clinical medicine ,Immunology and Microbiology (miscellaneous) ,Models ,Site-Directed ,Induced pluripotent stem cell ,ALS ,FUS ,iPSCs ,TALE nucleases ,Active Transport, Cell Nucleus ,Amino Acid Substitution ,Amyotrophic Lateral Sclerosis ,Cell Differentiation ,Cell Line ,Cytoplasmic Granules ,Humans ,Induced Pluripotent Stem Cells ,Models, Neurological ,Motor Neurons ,Mutagenesis, Site-Directed ,Mutant Proteins ,RNA-Binding Protein FUS ,Spinal Cord ,Stress, Physiological ,Biochemistry, Genetics and Molecular Biology (all) ,Neuroscience (miscellaneous) ,0303 health sciences ,Mutation ,Phenotype ,Active Transport ,Neurological ,motoneurons ,lcsh:RB1-214 ,stress granules ,Physiological ,Mutagenesis (molecular biology technique) ,Biology ,Stress ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Stress granule ,lcsh:Pathology ,medicine ,Resource Article ,030304 developmental biology ,Cell Nucleus ,lcsh:R ,Molecular biology ,Mutagenesis ,030217 neurology & neurosurgery - Abstract
Patient-derived induced pluripotent stem cells (iPSCs) provide an opportunity to study human diseases mainly in those cases for which no suitable model systems are available. Here, we have taken advantage of in vitro iPSCs derived from patients affected by amyotrophic lateral sclerosis (ALS) and carrying mutations in the RNA-binding protein FUS to study the cellular behavior of the mutant proteins in the appropriate genetic background. Moreover, the ability to differentiate iPSCs into spinal cord neural cells provides an in vitro model mimicking the physiological conditions. iPSCs were derived from FUSR514S and FUSR521C patient fibroblasts, whereas in the case of the severe FUSP525L mutation, in which fibroblasts were not available, a heterozygous and a homozygous iPSC line were raised by TALEN-directed mutagenesis. We show that aberrant localization and recruitment of FUS into stress granules (SGs) is a prerogative of the FUS mutant proteins and occurs only upon induction of stress in both undifferentiated iPSCs and spinal cord neural cells. Moreover, we show that the incorporation into SGs is proportional to the amount of cytoplasmic FUS, strongly correlating with the cytoplasmic delocalization phenotype of the different mutants. Therefore, the available iPSCs represent a very powerful system for understanding the correlation between FUS mutations, the molecular mechanisms of SG formation and ALS ethiopathogenesis., Summary: Mutated FUS protein is aberrantly delocalized and recruited into stress granules in iPSC-derived motoneurons, which provide a new model system for amyotrophic lateral sclerosis.
- Published
- 2015
39. C/EBPa-p30 protein induces expression of the oncogenic long non-coding RNA UCA1 in acute myeloid leukemia
- Author
-
Ivano Legnini, Beatrice Salvatori, Mariangela Morlando, Francesco Fazi, Alessandro Fatica, James M. Hughes, Marcella Marchioni, Silvia Masciarelli, and Irene Bozzoni
- Subjects
Myeloid ,lncRN ,CEBPA, AML ,long non-coding RNA ,acute myeloid leukemia ,CEBPA ,UCA1 ,medicine.disease_cause ,AML ,RNA interference ,Protein Isoforms ,Promoter Regions, Genetic ,Genetics ,Leukemic ,Tumor ,Leukemia ,Gene Expression Regulation, Leukemic ,Reverse Transcriptase Polymerase Chain Reaction ,Myeloid leukemia ,Cell cycle ,Long non-coding RNA ,3. Good health ,medicine.anatomical_structure ,Oncology ,Leukemia, Myeloid ,Acute Disease ,RNA, Long Noncoding ,RNA Interference ,Long Noncoding ,Settore BIO/17 - ISTOLOGIA ,Acute myeloid leukemia ,Biomarkers, Tumor ,CCAAT-Enhancer-Binding Protein-alpha ,Cell Proliferation ,Cyclin-Dependent Kinase Inhibitor p27 ,Humans ,Immunoblotting ,K562 Cells ,Mutation ,Protein Binding ,Gene Expression Profiling ,Research Paper ,Promoter Regions ,Genetic ,medicine ,business.industry ,Gene expression profiling ,Gene Expression Regulation ,Cancer research ,RNA ,business ,Carcinogenesis ,Biomarkers - Abstract
// James M. Hughes 1 , Ivano Legnini 1 , Beatrice Salvatori 1, 6 , Silvia Masciarelli 2 , Marcella Marchioni 3 , Francesco Fazi 2 , Mariangela Morlando 1 , Irene Bozzoni 1, 3, 4, 5 , Alessandro Fatica 1 1 Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy 2 Department of Anatomical, Histological, Forensic & Orthopaedic Sciences, Sapienza University of Rome, Rome, Italy 3 Institute of Biology, Molecular Medicine and Nanobiotechnology, CNR, Sapienza University of Rome, Rome, Italy 4 Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy 5 Institute Pasteur Fondazione Cenci-Bolognetti, Sapienza University of Rome, Rome, Italy 6 Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA Correspondence to: Alessandro Fatica, e-mail: alessandro.fatica@uniroma1.it Keywords: long non-coding RNA, acute myeloid leukemia, CEBPA, UCA1 Received: February 05, 2015 Accepted: May 13, 2015 Published: May 25, 2015 ABSTRACT Accumulating evidences indicate that different long non-coding RNAs (lncRNAs) might play a relevant role in tumorigenesis, with their expression and function already associated to cancer development and progression. CCAAT/enhancer-binding protein-α (CEBPA) is a critical regulator of myeloid differentiation whose inactivation contributes to the development of acute myeloid leukemia (AML). Mutations in C/EBPα occur in around 10% of AML cases, leading to the expression of a 30-kDa dominant negative isoform (C/EBPα-p30). In this study, we identified the oncogenic urothelial carcinoma associated 1 (UCA1) lncRNA as a novel target of the C/EBPα-p30. We show that wild-type C/EBPα and C/EBPα-p30 isoform can bind the UCA1 promoter but have opposite effects on UCA1 expression. While wild-type C/EBPα represses, C/EBPα-p30 can induce UCA1 transcription. Notably, we also show that UCA1 expression increases in cytogenetically normal AML cases carrying biallelic CEBPA mutations. Furthermore, we demonstrate that UCA1 sustains proliferation of AML cells by repressing the expression of the cell cycle regulator p27 kip1 . Thus, we identified, for the first time, an oncogenic lncRNA functioning in concert with the dominant negative isoform of C/EBPα in AML.
- Published
- 2015
40. Long Non-Coding RNAs: New Players in Hematopoiesis and Leukemia
- Author
-
Mariangela Morlando, Monica Ballarino, and Alessandro Fatica
- Subjects
hematopoiesis ,lncRNA ,leukemia ,Bioinformatics ,Blood cell ,Transcription (biology) ,Gene expression ,medicine ,cancer ,Gene ,lcsh:R5-920 ,Mini Reviews in Medicine ,business.industry ,RNA ,General Medicine ,medicine.disease ,3. Good health ,Cell biology ,Leukemia ,Haematopoiesis ,medicine.anatomical_structure ,RNA splicing ,Medicine ,blood cells ,business ,lcsh:Medicine (General) - Abstract
Long non-coding RNAs (lncRNAs) are important regulators of gene expression that influence almost every step in the life cycle of genes, from transcription to mRNA splicing, RNA decay, and translation. Besides their participation to normal physiology, lncRNA expression and function have been already associated to cancer development and progression. Here, we review the functional role and mechanisms of action of lncRNAs in normal hematopoiesis and how their misregulation may be implicated in the development of blood cell cancer, such as leukemia.
- Published
- 2015
41. Novel long noncoding RNAs (lncRNAs) in Myogenesis: A miR-31 overlapping lncRNA transcript controls myoblast differentiation
- Author
-
Tiziana Santini, Chiara Pinnarò, Irene Bozzoni, Valentina Cazzella, Lavinia Bisceglie, Anna Tramontano, Mariangela Morlando, Daniel D'Andrea, Monica Ballarino, Andrea Cipriano, and Luigi Grassi
- Subjects
Duchenne muscular dystrophy ,Cellular differentiation ,Biology ,Muscle Development ,Transgenic ,Transcriptome ,Striated ,Myoblasts ,Mice ,microRNA ,medicine ,RNA Precursors ,Animals ,Humans ,Muscular Dystrophy ,Molecular Biology ,Biological Evolution ,Cell Differentiation ,Cell Proliferation ,Gene Expression Profiling ,Gene Expression Regulation ,Mice, Transgenic ,MicroRNAs ,Muscle, Smooth ,Muscle, Striated ,Muscular Dystrophy, Duchenne ,RNA, Long Noncoding ,Cell Biology ,Regulation of gene expression ,Myogenesis ,RNA ,medicine.disease ,Duchenne ,Molecular biology ,Gene expression profiling ,Muscle ,Long Noncoding ,Smooth - Abstract
Transcriptome analysis allowed the identification of new long noncoding RNAs differentially expressed during murine myoblast differentiation. These transcripts were classified on the basis of their expression under proliferating versus differentiated conditions, muscle-restricted activation, and subcellular localization. Several species displayed preferential expression in dystrophic (mdx) versus wild-type muscles, indicating their possible link with regenerative processes. One of the identified transcripts, lnc-31, even if originating from the same nuclear precursor of miR-31, is produced by a pathway mutually exclusive. We show that lnc-31 and its human homologue hsa-lnc-31 are expressed in proliferating myoblasts, where they counteract differentiation. In line with this, both species are more abundant in mdx muscles and in human Duchenne muscular dystrophy (DMD) myoblasts, than in their normal counterparts. Altogether, these data suggest a crucial role for lnc-31 in controlling the differentiation commitment of precursor myoblasts and indicate that its function is maintained in evolution despite the poor sequence conservation with the human counterpart.
- Published
- 2015
42. MicroRNAs and Hematopoietic Differentiation
- Author
-
Alessandro Rosa, Alessandro Fatica, Clara Nervi, Elisa Caffarelli, F. De Angelis, Francesco Fazi, Monica Ballarino, Irene Bozzoni, and Mariangela Morlando
- Subjects
Cellular differentiation ,Cell Differentiation ,genetics/physiology, Gene Expression Regulation ,Developmental, Hematopoiesis ,genetics/physiology, Humans, Leukemia ,genetics/metabolism, MicroRNAs ,genetics/metabolism, Models ,Biological, RNA ,Neoplasm ,genetics/metabolism, Transcription Factors ,metabolism ,Computational biology ,Biology ,Models, Biological ,Biochemistry ,Gene Expression Regulation, Developmental ,Hematopoiesis ,Humans ,Leukemia ,MicroRNAs ,RNA, Neoplasm ,Transcription Factors ,Models ,Gene expression ,microRNA ,Genetics ,Developmental ,Molecular Biology ,Transcription factor ,Gene ,Alternative splicing ,Translation (biology) ,Biological ,Gene Expression Regulation ,RNA ,Human genome - Abstract
The discovery of microRNAS (miRNAs) and of their mechanism of action has provided some very new clues on how gene expression is regulated. These studies established new concepts on how posttranscriptional control can fine-tune gene expression during differentiation and allowed the identification of new regulatory circuitries as well as factors involved therein. Because of the wealth of information available about the transcriptional and cellular networks involved in hematopoietic differentiation, the hematopoietic system is ideal for studying cell lineage specification. An interesting interplay between miRNAs and lineage-specific transcriptional factors has been found, and this can help us to understand how terminal differentiation is accomplished.
- Published
- 2006
43. Functional Analysis of Yeast snoRNA and snRNA 3′-End Formation Mediated by Uncoupling of Cleavage and Polyadenylation
- Author
-
Walter Keller, Alessandro Fatica, Bernhard Dichtl, Paolo Greco, Mariangela Morlando, and Irene Bozzoni
- Subjects
Cleavage factor ,Saccharomyces cerevisiae Proteins ,Polyadenylation ,Genes, Fungal ,Molecular Sequence Data ,Gene Expression ,Saccharomyces cerevisiae ,Cleavage and polyadenylation specificity factor ,Biology ,Cleavage (embryo) ,Chromosomes ,Fungal Proteins ,Small Nuclear ,RNA, Small Nuclear ,RNA Precursors ,RNA, Small Nucleolar ,Small nucleolar RNA ,Base Sequence ,Chromosomes, Fungal ,Mutation ,Nuclear Proteins ,RNA, Fungal ,RNA 3' End Processing ,mRNA Cleavage and Polyadenylation Factors ,Molecular Biology ,Small Nucleolar ,Genetics ,Cleavage stimulation factor ,Fungal genetics ,Cell Biology ,Fungal ,Genes ,RNA ,Small nuclear RNA - Abstract
Many nuclear and nucleolar small RNAs are accumulated as nonpolyadenylated species and require 3'-end processing for maturation. Here, we show that several genes coding for box C/D and H/ACA snoRNAs and for the U5 and U2 snRNAs contain sequences in their 3' portions which direct cleavage of primary transcripts without being polyadenylated. Genetic analysis of yeasts with mutations in different components of the pre-mRNA cleavage and polyadenylation machinery suggests that this mechanism of 3"-end formation requires cleavage factor IA (CF IA) but not cleavage and polyadenylation factor activity. However, in vitro results indicate that other factors participate in the reaction besides CF IA. Sequence analysis of snoRNA genes indicated that they contain conserved motifs in their 3" noncoding regions, and mutational studies demonstrated their essential role in 3"-end formation. We propose a model in which CF IA functions in cleavage and polyadenylation of pre-mRNAs and, in combination with a different set of factors, in 3"-end formation of nonpolyadenylated polymerase II transcripts.
- Published
- 2002
44. Circ-ZNF609 Is a Circular RNA that Can Be Translated and Functions in Myogenesis
- Author
-
Tiziana Santini, Irene Bozzoni, Gaia Di Timoteo, Alessandro Fatica, Adrian Andronache, Mark Wade, Mariangela Morlando, Francesca Rossi, Francesca Briganti, Ivano Legnini, Pietro Laneve, Olga Sthandier, and Nikolaus Rajewsky
- Subjects
Male ,0301 basic medicine ,Myoblast proliferation ,non-coding RNA ,translation ,Muscle Proteins ,Muscle Development ,Myoblasts ,Mice ,Start codon ,circRNA ,Muscular Dystrophy ,Genetics ,Myogenesis ,High-Throughput Nucleotide Sequencing ,Skeletal ,Non-coding RNA ,Cell biology ,Phenotype ,RNA splicing ,RNA Interference ,myogenesis ,Sequence Analysis ,Signal Transduction ,RNA Caps ,Genotype ,proliferation ,RNA Splicing ,muscle differentiation ,cap independent ,circular RNA ,DMD ,Molecular Biology ,Cell Biology ,Biology ,Transfection ,Article ,Open Reading Frames ,03 medical and health sciences ,Circular RNA ,Polysome ,Animals ,Humans ,Cell Proliferation ,HeLa Cells ,Muscular Dystrophy, Duchenne ,Myoblasts, Skeletal ,RNA ,Sequence Analysis, RNA ,Protein Biosynthesis ,Duchenne ,Open reading frame ,030104 developmental biology ,Cardiovascular and Metabolic Diseases - Abstract
Summary Circular RNAs (circRNAs) constitute a family of transcripts with unique structures and still largely unknown functions. Their biogenesis, which proceeds via a back-splicing reaction, is fairly well characterized, whereas their role in the modulation of physiologically relevant processes is still unclear. Here we performed expression profiling of circRNAs during in vitro differentiation of murine and human myoblasts, and we identified conserved species regulated in myogenesis and altered in Duchenne muscular dystrophy. A high-content functional genomic screen allowed the study of their functional role in muscle differentiation. One of them, circ-ZNF609, resulted in specifically controlling myoblast proliferation. Circ-ZNF609 contains an open reading frame spanning from the start codon, in common with the linear transcript, and terminating at an in-frame STOP codon, created upon circularization. Circ-ZNF609 is associated with heavy polysomes, and it is translated into a protein in a splicing-dependent and cap-independent manner, providing an example of a protein-coding circRNA in eukaryotes., Graphical Abstract, Highlights • CircRNAs are conserved, abundant, and regulated in myogenesis • High-throughput phenotypic screening reveals functional circRNAs • Circ-ZNF609 regulates myoblast proliferation • Circ-ZNF609 can be translated, Legnini et al. identified circ-ZNF609, a circular RNA expressed in murine and human myoblasts, which controls myoblast proliferation. Circ-ZNF609 contains an open reading frame and is translated into a protein in a splicing-dependent/cap-independent manner. Circ-ZNF609 translation can be modulated by stress conditions.
- Published
- 2017
45. The role of long noncoding RNAs in the epigenetic control of gene expression
- Author
-
Irene Bozzoni, Alessandro Fatica, Monica Ballarino, and Mariangela Morlando
- Subjects
Heterochromatin ,Computational biology ,Biochemistry ,Epigenesis, Genetic ,Transcriptome ,long noncoding rna ,epigenetics ,histones ,modular scaffolds ,chromatin ,Genetic ,Drug Discovery ,Gene expression ,long noncoding RNA ,Epigenetics ,General Pharmacology, Toxicology and Pharmaceutics ,Pharmacology ,Genetics ,biology ,Organic Chemistry ,RNA, Long Noncoding ,RNA ,Long non-coding RNA ,Chromatin ,Histone ,biology.protein ,Molecular Medicine ,Long Noncoding ,Epigenesis - Abstract
Recent advances in the methodologies employed to deeply analyse the complexity of transcriptomes have unveiled the existence of a new class of transcripts, long noncoding RNAs (lncRNAs). A significant amount of effort has been dedicated to the study of lncRNAs, and a large body of evidence now exists indicating their relevant role in different regulatory steps of gene expression. Given the role of epigenetics in disease development and progression, this Minireview focuses on lncRNAs involved in epigenetic control and provides an overview of the mechanisms used to guide epigenetic-modifying complexes to adjacent (cis-acting) or independent (trans-acting) genomic loci. Furthermore, it describes the activities of these transcripts in controlling the formation and spreading of heterochromatin domains. Just as other RNA molecules have found therapeutic application, though much remains to be elucidated about the structure and function of these lncRNAs, they too could hold potential as biomarkers, targets, and therapeutic agents.
- Published
- 2014
46. An ALS-associated mutation in the FUS 3'-UTR disrupts a microRNA-FUS regulatory circuitry
- Author
-
Mario Sabatelli, Stefano Dini Modigliani, Lorenzo Errichelli, Irene Bozzoni, and Mariangela Morlando
- Subjects
Untranslated region ,sclerosi laterale amiotrofica ,microrna ,General Physics and Astronomy ,Biology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Pathogenesis ,fus ,microRNA ,medicine ,Humans ,Amyotrophic lateral sclerosis ,3' Untranslated Regions ,Regulation of gene expression ,Homeodomain Proteins ,Mutation ,Multidisciplinary ,Three prime untranslated region ,Amyotrophic Lateral Sclerosis ,Zinc Finger E-box-Binding Homeobox 1 ,General Chemistry ,Regulatory loop ,medicine.disease ,MicroRNAs ,Gene Expression Regulation ,Cancer research ,RNA-Binding Protein FUS ,Transcription Factors - Abstract
While the physiologic functions of the RNA-binding protein FUS still await thorough characterization, the pathonegetic role of FUS mutations in amyotrophic lateral sclerosis (ALS) is clearly established. Here we find that a human FUS mutation that leads to increased protein expression, and was identified in two ALS patients with severe outcome, maps to the seed sequence recognized by miR-141 and miR-200a in the 3'-UTR of FUS. We demonstrate that FUS and these microRNAs are linked by a feed-forward regulatory loop where FUS upregulates miR-141/200a, which in turn impact FUS protein synthesis. We also show that Zeb1, a target of miR-141/200a and transcriptional repressor of these two microRNAs, is part of the circuitry and reinforces it. Our results reveal a possible correlation between deregulation of this regulatory circuit and ALS pathogenesis, and open interesting perspectives in the treatment of these mutations through ad hoc-modified microRNAs.
- Published
- 2014
47. Effects of FUS/TLS and its mutations on the in vitro properties of Neural Progenitor cell lines and their neural progeny
- Author
-
Camilla, Toselli, Mariangela, Morlando, Bozzoni, Irene, Biagioni, Stefano, and Cacci, Emanuele
- Subjects
FUS/TLS, Neural stem cells, differentiation ,Neural stem cells ,differentiation ,FUS/TLS - Published
- 2013
48. Biogenesis and function of non-coding RNAs in muscle differentiation and in Duchenne muscular dystrophy
- Author
-
Ivano Legnini, Shyam Sundar Twayana, Irene Bozzoni, Mariangela Morlando, Marcella Cesana, Davide Cacchiarelli, Twayana, Shyam, Legnini, Ivano, Cesana, Marcella, Cacchiarelli, Davide, Morlando, Mariangela, and Bozzoni, Irene
- Subjects
Duchenne muscular dystrophy ,Long non-coding RNA (lncRNA) ,MicroRNA (miRNA) ,Muscle differentiation ,Cell Differentiation ,Humans ,MicroRNAs ,Muscle, Skeletal ,Muscular Dystrophy, Duchenne ,RNA, Untranslated ,duchenne muscular dystrophy ,long non coding rna ,long non-coding rna (incrna) ,long non-coding rna (lncrna) ,microrna ,microrna (mirna) ,muscle differentiation ,Cellular differentiation ,Computational biology ,Biology ,Biochemistry ,Transcriptome ,microRNA ,Muscular Dystrophy ,Genetics ,Competing endogenous RNA ,RNA ,Untranslated ,MicroRNA ,Skeletal ,Duchenne ,Long non-coding RNA ,Muscle ,Function (biology) ,Biogenesis ,Human - Abstract
It is now becoming largely accepted that the non-coding portion of the genome, rather than its coding counterpart, is likely to account for the greater complexity of higher eukaryotes. Moreover, non-coding RNAs have been demonstrated to participate in regulatory circuitries that are crucial for development and differentiation. Whereas the biogenesis and function of small non-coding RNAs, particularly miRNAs (microRNAs), has been extensively clarified in many eukaryotic systems, very little is known about the long non-coding counterpart of the transcriptome. In the present review, we revise the current knowledge of how small non-coding RNAs and lncRNAs (long non-coding RNAs) impinge on circuitries controlling proper muscle differentiation and homoeostasis and how their biogenesis is regulated. Moreover, we provide new insights into an additional mechanism of post-transcriptional regulation mediated by lncRNAs, which, acting as miRNA ‘sponges’, have an impact on the distribution of miRNA molecules on their targets with features similar to those described for ceRNAs (competing endogenous RNAs).
- Published
- 2013
49. MicroRNAs involved in molecular circuitries relevant for the duchenne muscular dystrophy pathogenesis are controlled by the dystrophin/nNOS pathway
- Author
-
Alberto Auricchio, Laura Barberi, Tiziana Santini, Mariangela Morlando, Olga Sthandier, Carmine Nicoletti, Tania Incitti, Erika Girardi, Julie Martone, Antonio Musarò, Irene Bozzoni, Marcella Cesana, Davide Cacchiarelli, Cacchiarelli, Davide, Martone, Julie, Girardi, Erika, Cesana, Marcella, Incitti, Tania, Morlando, Mariangela, Nicoletti, Carmine, Santini, Tiziana, Sthandier, Olga, Barberi, Laura, Auricchio, Alberto, Musar, Antonio, and Bozzoni, Irene
- Subjects
Physiology ,Duchenne muscular dystrophy ,HUMDISEASE ,Histone Deacetylase 2 ,DEVBIO ,Nitric Oxide Synthase Type I ,Dystrophin ,Mice ,0302 clinical medicine ,Muscular Dystrophy ,Muscular dystrophy ,Regulation of gene expression ,0303 health sciences ,Nitrosylation ,MicroRNA ,Skeletal ,musculoskeletal system ,Chromatin ,Cell biology ,Nitric oxide synthase ,Satellite Cells ,Muscle ,musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,Skeletal Muscle ,Satellite Cells, Skeletal Muscle ,DNA ,Animals ,Gene Expression Regulation ,Mice, Inbred mdx ,MicroRNAs ,Muscle, Skeletal ,Muscular Dystrophy, Animal ,Regeneration ,Biology ,03 medical and health sciences ,medicine ,devbio ,dna ,humdisease ,Molecular Biology ,030304 developmental biology ,Sarcolemma ,Animal ,Inbred mdx ,Cell Biology ,medicine.disease ,biology.protein ,Cancer research ,030217 neurology & neurosurgery - Abstract
SummaryIn Duchenne muscular dystrophy (DMD) the absence of dystrophin at the sarcolemma delocalizes and downregulates nitric oxide synthase (nNOS); this alters S-nitrosylation of HDAC2 and its chromatin association. We show that the differential HDAC2 nitrosylation state in Duchenne versus wild-type conditions deregulates the expression of a specific subset of microRNA genes. Several circuitries controlled by the identified microRNAs, such as the one linking miR-1 to the G6PD enzyme and the redox state of cell, or miR-29 to extracellular proteins and the fibrotic process, explain some of the DMD pathogenetic traits. We also show that, at variance with other myomiRs, miR-206 escapes from the dystrophin-nNOS control being produced in activated satellite cells before dystrophin expression; in these cells, it contributes to muscle regeneration through repression of the satellite specific factor, Pax7. We conclude that the pathway activated by dystrophin/nNOS controls several important circuitries increasing the robustness of the muscle differentiation program.
- Published
- 2010
50. Coupling between snoRNP assembly and 3′ processing controls box C/D snoRNA biosynthesis in yeast
- Author
-
Irene Bozzoni, Paolo Greco, Elisa Caffarelli, Monica Ballarino, Mariangela Morlando, and Bernhard Dichtl
- Subjects
3′-end formation ,Nop1p ,Saccharomyces cerevisiae ,snoRNA ,snoRNP assembly ,Base Sequence ,DNA Primers ,Mutation ,Plasmids ,Ribonucleoproteins, Small Nucleolar ,Polyadenylation ,RNA polymerase II ,Cleavage (embryo) ,General Biochemistry, Genetics and Molecular Biology ,Article ,Transcription (biology) ,Small nucleolar RNA ,Molecular Biology ,Gene ,Base Sequence, DNA Primers, Mutation, Plasmids, Ribonucleoproteins ,Small Nucleolar ,metabolism, Saccharomyces cerevisiae ,metabolism ,Genetics ,General Immunology and Microbiology ,biology ,urogenital system ,General Neuroscience ,biology.organism_classification ,Cell biology ,Ribonucleoproteins ,biology.protein ,Small nuclear RNA - Abstract
RNA polymerase II transcribes genes encoding proteins and a large number of small stable RNAs. While pre-mRNA 3′-end formation requires a machinery ensuring tight coupling between cleavage and polyadenylation, small RNAs utilize polyadenylation-independent pathways. In yeast, specific factors required for snRNA and snoRNA 3′-end formation were characterized as components of the APT complex that is associated with the core complex of the cleavage/polyadenylation machinery (core-CPF). Other essential factors were identified as independent components: Nrd1p, Nab3p and Sen1p. Here we report that mutations in the conserved box D of snoRNAs and in the snoRNP-specific factor Nop1p interfere with transcription and 3′-end formation of box C/D snoRNAs. We demonstrate that Nop1p is associated with box C/D snoRNA genes and that it interacts with APT components. These data suggest a mechanism of quality control in which efficient transcription and 3′-end formation occur only when nascent snoRNAs are successfully assembled into functional particles.
- Published
- 2004
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.