102 results on '"Persichetti F"'
Search Results
2. Recurrent simple tandem repeat mutations during human Y-chromosome radiation in Caucasian subpopulations
- Author
-
Ciminelli, B. M., Pompei, F., Malaspina, P., Hammer, M., Persichetti, F., Pignatti, P. F., Palena, A., Anagnou, N., Guanti, G., Jodice, C., Terrenato, L., and Novelletto, A.
- Published
- 1995
- Full Text
- View/download PDF
3. RRS1 is involved in endoplasmic reticulum stress response in Huntington's disease
- Author
-
Carnemolla A., Fossale E., Agostoni E., Michelazzi S., Calligaris R., De Maso L., Macdonald M., Persichetti F., DEL SAL, GIANNINO, Carnemolla, A., Fossale, E., Agostoni, E., Michelazzi, S., Calligaris, R., De Maso, L., DEL SAL, Giannino, Macdonald, M., and Persichetti, F.
- Published
- 2009
4. SINEUPs: A new class of natural and synthetic antisense long non-coding RNAs that activate translation
- Author
-
Zucchelli, S, primary, Cotella, D, additional, Takahashi, H, additional, Carrieri, C, additional, Cimatti, L, additional, Fasolo, F, additional, Jones, MH, additional, Sblattero, D, additional, Sanges, R, additional, Santoro, C, additional, Persichetti, F, additional, Carninci, P, additional, and Gustincich, S, additional
- Published
- 2015
- Full Text
- View/download PDF
5. The gene for autosomal dominant spinocerebellar ataxia (SCA1) maps centromeric to D6S89 and shows no recombination, in nine large kindreds, with a dinucleotide repeat at the AM10 locus
- Author
-
Kwiatkowski TJ, J.r., Orr HT, McCall AE, Jodice C, Persichetti F, Novelletto A, LeBorgne DeMarquoy F, Duvick LA, Frontali M. and, BANFI, Sandro, Kwiatkowski, Tj, J., R., Orr, Ht, Banfi, Sandro, Mccall, Ae, Jodice, C, Persichetti, F, Novelletto, A, LeBorgne DeMarquoy, F, Duvick, La, and Frontali M., And
- Published
- 1993
6. Effect of trinucleotide repeat length and parental sex on phenotypic variation in spinocerebellar ataxia I
- Author
-
Jodice, C., Malaspina, P., Persichetti, F., Novelletto, A., Spadaro, M., Paola Giunti, Morocutti, C., Terrenato, L., Harding, Ae, and Frontali, M.
- Subjects
Adult ,Male ,Parents ,Polymorphism, Genetic ,Base Sequence ,Molecular Sequence Data ,Middle Aged ,Phenotype ,Sex Factors ,Oligodeoxyribonucleotides ,Humans ,Female ,Research Article ,Repetitive Sequences, Nucleic Acid ,Spinocerebellar Degenerations - Abstract
Trinucleotide repeat expansion has been found in 64 subjects from 19 families: 57 patients with SCA1 and 7 subjects predicted, by haplotype analysis, to carry the mutation. Comparison with a large set of normal chromosomes shows two distinct distributions, with a much wider variation among expanded chromosomes. The sex of transmitting parent plays a major role in the size distribution of expanded alleles, those with > 54 repeats being transmitted by affected fathers exclusively. Our data suggest that alleles with > 54 repeats have a reduced chance of survival; these appear to be replaced in each generation by further expansion of alleles in the low- to medium-expanded repeat range, preferentially in male transmissions. Detailed clinical follow-up of a subset of our patients demonstrates significant relationships between increasing repeat number on expanded chromosomes and earlier age at onset, faster progression of the disease, and earlier age at death.
- Published
- 1994
7. Polymorphism analysis of the huntingtin gene in Italian families affected with Huntington disease
- Author
-
Novelletto, A., Persichetti, F., Sabbadini, G., Mandich, Paola, Bellone, Emilia, Ajmar, Franco, Squitieri, F., Campanella, G., Bozza, A., and Macdonald, M. E.
- Subjects
Adult ,epidemiology/genetics ,Adult, Age of Onset, Codon ,genetics, Genes, Humans, Huntington Disease ,epidemiology/genetics, Italy ,epidemiology, Linkage Disequilibrium, Middle Aged, Minisatellite Repeats, Nerve Tissue Proteins ,genetics, Nuclear Proteins ,genetics, Polymerase Chain Reaction, Polymorphism ,Genetic, Sequence Deletion ,Nuclear Proteins ,Nerve Tissue Proteins ,Minisatellite Repeats ,Middle Aged ,Polymerase Chain Reaction ,Linkage Disequilibrium ,Huntington Disease ,Genes ,Italy ,Genetic ,Humans ,genetics ,epidemiology ,Age of Onset ,Polymorphism ,Codon ,Sequence Deletion - Published
- 1994
8. The Gene for Spinal Cerebellar Ataxia 1 is Flanked by Two Closely Linked Highly Polymorphic Microsatellite Loci
- Author
-
Jodice, C, Frontali, M, Persichetti, F, Novelletto, A, Pandolfo, M, Spadaro, M, Giunti, P, Schinaia, Giuseppe, Lulli, Patrizia, Malaspina, P, Plasmati, R, Tola, R, Antonelli, A, DI DONATO, S, Morocutti, C, Weissenbach, J, Cann, Hm, and Terrenato, L.
- Published
- 1993
9. HLA-linked spinocerebellar ataxia: a clinical and genetic study of large Italian kindreds
- Author
-
Spadaro, M., primary, Giunti, P., additional, Lulli, P., additional, Frontali, M., additional, Jodice, C., additional, Cappellacci, S., additional, Morellini, M., additional, Persichetti, F., additional, Trabace, S., additional, Anastasi, R., additional, and Morocutti, C., additional
- Published
- 2009
- Full Text
- View/download PDF
10. Linkage analysis and linkage disequilibrium studies between SCA1 and TG microsatellite VNTRs on 6p
- Author
-
Iodice, C, Persichetti, F, Frontali, M, Lulli, P, Spadaro, M, Cappellacci, S, Giunti, P, Malaspina, P, Morellini, M, Morocutti, C, Novelletto, A, Trabace, S, and Terrenato, L
- Subjects
Settore BIO/18 - Genetica ,Settore MED/03 - Genetica Medica - Published
- 1991
11. SPINOCEREBELLAR ATAXIA (SCA1) IN 2 LARGE ITALIAN KINDREDS - EVIDENCE IN FAVOR OF A LOCUS POSITION DISTAL TO GLO1 AND THE HLA CLUSTER
- Author
-
Frontali, M, Iodice, C, Lulli, P, Spadaro, M, Cappellacci, S, Giunti, P, Malaspina, P, Morellini, M, Morocutti, C, Novelletto, A, Persichetti, F, Trabace, S, Anastasi, R, and Terrenato, L
- Subjects
F13A ,HEREDITARY ATAXIA ,Settore BIO/18 - Genetica ,MULTILOCUS LINKAGE ANALYSIS ,HUMAN CHROMOSOME-6 ,CEREBELLAR-ATAXIA ,FACTOR-XIIIA ,SHORT ARM ,POLYMORPHISM ,POPULATION ,ONSET - Published
- 1991
12. The gene for spinal cerebellar ataxia 1 (SCA1) is flanked by two closely linked highly polymorphic microsatellite loci.
- Author
-
Jodice, C, Frontali, M, Persichetti, F, Novelletto, A, Pandolfo, Massimo, Spadaro, M, Giunti, P, Schinaia, G, Lulli, P, Malaspina, P, Jodice, C, Frontali, M, Persichetti, F, Novelletto, A, Pandolfo, Massimo, Spadaro, M, Giunti, P, Schinaia, G, Lulli, P, and Malaspina, P
- Abstract
The gene for one form of autosomal dominant spinal cerebellar ataxia (SCA1), is mapped by linkage to chromosome 6p, very close to the microsatellite locus D6S89. Eight large Italian kindreds segregating SCA1, as defined by very close linkage to D6S89, were genotyped with five microsatellite markers linked closely to D6S89, all mapping within a 6 cM interval on 6p. Multipoint linkage analysis and haplotypes from recombinants map SCA1 between two of these markers, D6S274 and D6S259, 5-6 cM apart. A single rare four marker haplotype within this interval shows linkage disequilibrium with the disease locus in southern Italy and is transmitted with SCA1 in five kindreds originating from this area., Journal Article, Research Support, Non-U.S. Gov't, info:eu-repo/semantics/published
- Published
- 1993
13. CEPH Consortium Map of Chromosome 14
- Author
-
Cox, D.W., primary, Billingsley, G.D., additional, Bale, A.E., additional, Donis-Keller, H., additional, Edwards, J.H., additional, Litt, M., additional, Mcbride, W., additional, Persichetti, F., additional, Spurr, N.K., additional, Weber, J.L., additional, Weissenbach, J., additional, and White, R.L., additional
- Published
- 1995
- Full Text
- View/download PDF
14. Analysis of the trinucleotide repeat expansion in Italian families affected with Huntington disease
- Author
-
Novelletto, A., primary, Persichetti, F., additional, Sabbadin, G., additional, Mandich, P., additional, Bellone, E., additional, Ajmar, F., additional, Pergola, M., additional, Senno, L. D., additional, E.MacDonald, M., additional, F.Gusella, J., additional, and Frontall, M., additional
- Published
- 1994
- Full Text
- View/download PDF
15. Trinucleotide repeat length instability and age of onset in Huntington's disease
- Author
-
Duyao, M., primary, Ambrose, C., additional, Myers, R., additional, Novelletto, A., additional, Persichetti, F., additional, Frontali, M., additional, Folstein, S., additional, Ross, C., additional, Franz, M., additional, Abbott, M., additional, Gray, J., additional, Conneally, P., additional, Young, A., additional, Penney, J., additional, Hollingsworth, Z., additional, Shoulson, I., additional, Lazzarini, A., additional, Falek, A., additional, Koroshetz, W., additional, Sax, D., additional, Bird, E., additional, Vonsattel, J., additional, Bonilla, E., additional, Alvir, J., additional, Bickham Conde, J., additional, Cha, J.-H., additional, Dure, L., additional, Gomez, F., additional, Ramos, M., additional, Sanchez-Ramos, J., additional, Snodgrass, S., additional, de Young, M., additional, Wexler, N., additional, Moscowitz, C., additional, Penchaszadeh, G., additional, MacFarlane, H., additional, Anderson, M., additional, Jenkins, B., additional, Srinidhi, J., additional, Barnes, G., additional, Gusella, J., additional, and MacDonald, M., additional
- Published
- 1993
- Full Text
- View/download PDF
16. AUTOSOMAL DOMINANT PURE CEREBELLAR ATAXIA
- Author
-
FRONTALI, M., primary, SPADARO, M., additional, GIUNTI, P., additional, BIANCO, F., additional, JODICE, C., additional, PERSICHETTI, F., additional, COLAZZA, G. B., additional, LULLI, P., additional, TERRENATO, L., additional, and MOROCUTTI, C., additional
- Published
- 1992
- Full Text
- View/download PDF
17. Spinocerebellar ataxia (SCA1) in two large Italian kindreds: evidence in favour of a locus position distal to GLO1 and the HLA cluster
- Author
-
FRONTALI, M., primary, IODICE, C., additional, LULLI, P., additional, SPADARO, M., additional, CAPPELLACCI, S., additional, GIUNTI, P., additional, MALASPINA, P., additional, MORELLINI, M., additional, MOROCUTTI, C., additional, NOVELLETTO, A., additional, PERSICHETTI, F., additional, TRABACE, S., additional, ANASTASI, R., additional, and TERRENATO, L., additional
- Published
- 1991
- Full Text
- View/download PDF
18. The human Y chromosome shows a low level of DNA polymorphism
- Author
-
MALASPINA, P., primary, PERSICHETTI, F., additional, NOVELLETTO, A., additional, IODICE, C., additional, TERRENATO, L., additional, WOLFE, J., additional, FERRARO, M., additional, and PRANTERA, G., additional
- Published
- 1990
- Full Text
- View/download PDF
19. An additional Hindlll polymorphism at the coagulation factor XlllA locus
- Author
-
Iodice, C., primary, Novelleto, A., additional, Malaspina, P., additional, and Persichetti, F., additional
- Published
- 1990
- Full Text
- View/download PDF
20. CEPH Consortium Map of Chromosome 14.
- Author
-
Cox, D.W., Billingsley, G.D., Bale, A.E., Donis-Keller, H., Edwards, J.H., Litt, M., Mcbride, W., Persichetti, F., Spurr, N.K., Weber, J.L., Weissenbach, J., and White, R.L.
- Published
- 1995
- Full Text
- View/download PDF
21. HLA-linked spinocerebellar ataxia: a clinical and genetic study of large Italian kindreds.
- Author
-
Spadaro, M., Giunti, P., Lulli, P., Frontali, M., Jodice, C., Cappellacci, S., Morellini, M., Persichetti, F., Trabace, S., Anastasi, R., and Morocutti, C.
- Published
- 1992
- Full Text
- View/download PDF
22. The gene for autosomal dominant spinocerebellar ataxia (SCAI) maps centromeric to D6S89 and shows no recombination, in nine large kindreds, with a dinucleotide repeat at the AM10 locus
- Author
-
Kwiatkowski Jr, T. J., Orr, H. T., Banfi, S., Mccall, A. E., Jodice, C., Persichetti, F., Andrea Novelletto, Leborgne-Demarquoy, F., Duvick, L. A., Frontali, M., Subramony, S. H., Beaudet, A. L., Terrenato, L., Zoghbi, H. Y., and Ranum, L. P. W.
23. Dominant phenotypes produced by the HD mutation in STHdh(Q111) striatal cells
- Author
-
flavia trettel, Rigamonti, D., Hilditch-Maguire, P., Wheeler, V. C., Sharp, A. H., Persichetti, F., Cattaneo, E., and Macdonald, M. E.
24. A human minisatellite hosts an alternative transcription start site for NPRL3 driving its expression in a repeat number-dependent manner
- Author
-
Raffaella Calligaris, Gilberto Pizzolato, Sara Finaurini, Piero Carninci, Lucia Antonutti, Remo Sanges, Maria Bertuzzi, Gianni Pezzoli, Dave Tang, Mauro Catalan, Stefano Goldwurm, Stefano Gustincich, Paolo Manganotti, Francesca Persichetti, Christina Vlachouli, Bertuzzi, M., Tang, D., Calligaris, R., Vlachouli, C., Finaurini, S., Sanges, R., Goldwurm, S., Catalan, M., Antonutti, L., Manganotti, P., Pizzolato, G., Pezzoli, G., Persichetti, F., Carninci, P., and Gustincich, S.
- Subjects
RNA Caps ,Genotype ,VNTR ,Minisatellite Repeats ,Biology ,Cell Line ,NPRL3 ,blood transcriptomic ,03 medical and health sciences ,Settore BIO/13 - Biologia Applicata ,blood transcriptomics ,minisatellite ,Gene expression ,Genetics ,Humans ,RNA, Small Interfering ,Gene ,Genetics (clinical) ,030304 developmental biology ,0303 health sciences ,Polymorphism, Genetic ,GTPase-Activating Proteins ,030305 genetics & heredity ,Intron ,Genomics ,Introns ,Cap analysis gene expression ,Variable number tandem repeat ,Minisatellite ,Gene Expression Regulation ,Multigene Family ,Expression quantitative trait loci ,RNA Interference ,Transcription Initiation Site - Abstract
Minisatellites, also called variable number of tandem repeats (VNTRs), are a class of repetitive elements that may affect gene expression at multiple levels and have been correlated to disease. Their identification and role as expression quantitative trait loci (eQTL) have been limited by their absence in comparative genomic hybridization and single nucleotide polymorphisms arrays. By taking advantage of cap analysis of gene expression (CAGE), we describe a new example of a minisatellite hosting a transcription start site (TSS) which expression is dependent on the repeat number. It is located in the third intron of the gene nitrogen permease regulator like protein 3 (NPRL3). NPRL3 is a component of the GAP activity toward rags 1 protein complex that inhibits mammalian target of rapamycin complex 1 (mTORC1) activity and it is found mutated in familial focal cortical dysplasia and familial focal epilepsy. CAGE tags represent an alternative TSS identifying TAGNPRL3 messenger RNAs (mRNAs). TAGNPRL3 is expressed in red blood cells both at mRNA and protein levels, it interacts with its protein partner NPRL2 and its overexpression inhibits cell proliferation. This study provides an example of a minisatellite that is both a TSS and an eQTL as well as identifies a new VNTR that may modify mTORC1 activity.
- Published
- 2020
25. The RNA-binding protein ILF3 binds to transposable element sequences in SINEUP lncRNAs
- Author
-
Francesca Fasolo, Remo Sanges, Piero Carninci, Francesca Persichetti, Flavio Mignone, Silvia Zucchelli, Claudio Santoro, Laura Patrucco, Clelia Peano, Carlotta Bon, Diego Cotella, Stefano Gustincich, Massimiliano Volpe, Daniele Sblattero, Fasolo, F., Patrucco, L., Volpe, M., Bon, C., Peano, C., Mignone, F., Carninci, P., Persichetti, F., Santoro, C., Zucchelli, S., Sblattero, D., Sanges, R., Cotella, D., and Gustincich, S.
- Subjects
0301 basic medicine ,Transposable element ,RNA-binding protein ,Computational biology ,Biology ,Biochemistry ,Domain (software engineering) ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Settore BIO/13 - Biologia Applicata ,Genetics ,Animals ,Humans ,Protein Interaction Domains and Motifs ,RNA, Antisense ,RNA, Messenger ,long noncoding RNA ,RIDome ,Nuclear Factor 90 Proteins ,Molecular Biology ,Research ,food and beverages ,Computational Biology ,Long non-coding RNA ,High-Throughput Screening Assays ,030104 developmental biology ,Protein Biosynthesis ,DNA Transposable Elements ,RNA, Long Noncoding ,Mammalian genome ,Ubiquitin Thiolesterase ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Transposable elements (TEs) compose about half of the mammalian genome and, as embedded sequences, up to 40% of long noncoding RNA (lncRNA) transcripts. Embedded TEs may represent functional domains within lncRNAs, providing a structured RNA platform for protein interaction. Here we show the interactome profile of the mouse inverted short interspersed nuclear element (SINE) of subfamily B2 (invSINEB2) alone and embedded in antisense (AS) ubiquitin C-terminal hydrolase L1 (Uchl1), an lncRNA that is AS to Uchl1 gene. AS Uchl1 is the representative member of a functional class of AS lncRNAs, named SINEUPs, in which the invSINEB2 acts as effector domain (ED)-enhancing translation of sense protein-coding mRNAs. By using RNA-interacting domainome technology, we identify the IL enhancer-binding factor 3 (ILF3) as a protein partner of AS Uchl1 RNA. We determine that this interaction is mediated by the RNA-binding motif 2 of ILF3 and the invSINEB2. Furthermore, we show that ILF3 is able to bind a free right Arthrobacter luteus (Alu) monomer sequence, the embedded TE acting as ED in human SINEUPs. Bioinformatic analysis of Encyclopedia of DNA Elements-enhanced cross-linking immunoprecipitation data reveals that ILF3 binds transcribed human SINE sequences at transcriptome-wide levels. We then demonstrate that the embedded TEs modulate AS Uchl1 RNA nuclear localization to an extent moderately influenced by ILF3. This work unveils the existence of a specific interaction between embedded TEs and an RNA-binding protein, strengthening the model of TEs as functional modules in lncRNAs.-Fasolo, F., Patrucco, L., Volpe, M., Bon, C., Peano, C., Mignone, F., Carninci, P., Persichetti, F., Santoro, C., Zucchelli, S., Sblattero, D., Sanges, R., Cotella, D., Gustincich, S. The RNA-binding protein ILF3 binds to transposable element sequences in SINEUP lncRNAs.
- Published
- 2019
26. Neuronal hemoglobin affects dopaminergic cells' response to stress
- Author
-
Paolo Ascenzi, Stefano Espinoza, Mauro Giacca, Margherita Francescatto, Roberta Russo, Daniela Cesselli, Lorena Zentilin, Francesca Persichetti, Marta Codrich, Giampiero Leanza, Stefano Gustincich, Silvia Zucchelli, Maria Bertuzzi, Antonio Paolo Beltrami, Codrich, M, Bertuzzi, M, Russo, R, Francescatto, M, Espinoza, S, Zentilin, L, Giacca, M, Cesselli, D, Beltrami, Ap, Ascenzi, Paolo, Zucchelli, S, Persichetti, F, Leanza, G, Gustincich, S., Marta, Codrich, Bertuzzi, Maria, Russo, Roberta, Francescatto, Margherita, Espinoza, Stefano, Zentilin, Lorena, Giacca, Mauro, Cesselli, Daniela, Beltrami, Antonio Paolo, Zucchelli, Silvia, Persichetti, Francesca, Leanza, Giampiero, and Stefano, Gustincich
- Subjects
0301 basic medicine ,MULTIPLE SYSTEM ATROPHY ,1-Methyl-4-phenylpyridinium ,Cancer Research ,Mouse ,ALPHA-SYNUCLEIN ,Dopamine ,Gene Expression ,Epigenesis, Genetic ,PARKINSONS-DISEASE ,GENE-EXPRESSION ,SH-SY5Y CELLS ,COMPLEX-I ,MICE ,AUTOPHAGY ,BRAINS ,TRANSCRIPTION ,Hemoglobins ,chemistry.chemical_compound ,0302 clinical medicine ,Settore BIO/13 - Biologia Applicata ,Viral Vector ,Parkinson's Disease ,MPTP ,Dopaminergic ,Brain ,Parkinson Disease ,Cell biology ,Substantia Nigra ,Biochemistry ,Original Article ,Hemoglobin ,Stress ,Autophagy ,Motor Learning ,medicine.drug ,Stre ,Immunology ,Substantia nigra ,Biology ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Neurochemical ,Rotenone ,Dopaminergic Cell ,medicine ,Animals ,Humans ,Parkinson Disease, Secondary ,Cell Biology ,Dopaminergic Neurons ,030104 developmental biology ,chemistry ,030217 neurology & neurosurgery ,Homeostasis - Abstract
Hemoglobin (Hb) is the major protein in erythrocytes and carries oxygen (O2) throughout the body. Recently, Hb has been found synthesized in atypical sites, including the brain. Hb is highly expressed in A9 dopaminergic (DA) neurons of the substantia nigra (SN), whose selective degeneration leads to Parkinson’s disease (PD). Here we show that Hb confers DA cells’ susceptibility to 1-methyl-4-phenylpyridinium (MPP+) and rotenone, neurochemical cellular models of PD. The toxic property of Hb does not depend on O2 binding and is associated with insoluble aggregate formation in the nucleolus. Neurochemical stress induces epigenetic modifications, nucleolar alterations and autophagy inhibition that depend on Hb expression. When adeno-associated viruses carrying α- and β-chains of Hb are stereotaxically injected into mouse SN, Hb forms aggregates and causes motor learning impairment. These results position Hb as a potential player in DA cells’ homeostasis and dysfunction in PD.
- Published
- 2017
27. Huntingtin polyQ Mutation Impairs the 17β-Estradiol/Neuroglobin Pathway Devoted to Neuron Survival
- Author
-
Pierangela Totta, Stefano Gustincich, Maria Marino, Mariarosa A. B. Melone, Paolo Ascenzi, Francesca Persichetti, Antonella Cardinale, Francesca Fusco, Maria Teresa Nuzzo, Marco Fiocchetti, Nuzzo, MARIA TERESA, Fiocchetti, Marco, Totta, Pierangela, Melone, Ma, Cardinale, Antonella, Fusco, Fr, Gustincich, S, Persichetti, F, Ascenzi, Paolo, Marino, Maria, Nuzzo, Mt, Fiocchetti, M, Totta, P, Melone, Mariarosa Anna Beatrice, Cardinale, A, Ascenzi, P, and Marino, M.
- Subjects
0301 basic medicine ,Huntingtin polyQ mutation ,congenital, hereditary, and neonatal diseases and abnormalities ,Huntingtin ,Cell Survival ,Neuroscience (miscellaneous) ,Neuroglobin ,Mice, Transgenic ,Nerve Tissue Proteins ,Biology ,Mitochondrion ,medicine.disease_cause ,Neuroprotection ,Hippocampus ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Mice ,0302 clinical medicine ,Huntington's disease ,Cell Line, Tumor ,mental disorders ,medicine ,Animals ,Neurons ,Mutation ,Huntingtin Protein ,Estradiol ,Neuron survival ,17β-estradiol, huntingtin, neuroglobin, Huntington’s disease, neuroprotective axis ,medicine.disease ,Corpus Striatum ,Globins ,Up-Regulation ,nervous system diseases ,17β-Estradiol ,030104 developmental biology ,Neurology ,nervous system ,Apoptosis ,Peptides ,Neuroscience ,030217 neurology & neurosurgery ,Function (biology) ,Signal Transduction - Abstract
Among several mechanisms underlying the well-known trophic and protective effects of 17β-estradiol (E2) in the brain, we recently reported that E2 induces the up-regulation of two anti-apoptotic and neuroprotectant proteins: huntingtin (HTT) and neuroglobin (NGB). Here, we investigate the role of this up-regulation. The obtained results indicate that E2 promotes NGB-HTT association, induces the localization of the complex at the mitochondria, and protects SK-N-BE neuroblastoma cells and murine striatal cells, which express wild-type HTT (i.e., polyQ7), against H2O2-induced apoptosis. All E2 effects were completely abolished in HTT-knocked out SK-N-BE cells and in striatal neurons expressing the mutated form of HTT (mHTT; i.e., polyQ111) typical of Huntington’s disease (HD). As a whole, these data provide a new function of wild-type HTT which drives E2-induced NGB in mitochondria modulating NGB anti-apoptotic activity. This new function is lost by HTT polyQ pathological expansion. These data evidence the existence of a novel E2/HTT/NGB neuroprotective axis that may play a relevant role in the development of HD therapeutics.
- Published
- 2017
28. Rrs1 Is Involved in Endoplasmic Reticulum Stress Response in Huntington Disease
- Author
-
Silvia Michelazzi, Giannino Del Sal, Francesca Persichetti, Elisa Fossale, Luca De Maso, Raffaella Calligaris, Alisia Carnemolla, Marcy E. MacDonald, Elena Agostoni, Carnemolla, A, Fossale, E, Agostoni, E, Michelazzi, S, Calligaris, Raffaella, De Maso, L, DEL SAL, Giannino, Macdonald, Me, and Persichetti, F.
- Subjects
Cytoplasm ,Nucleolus ,Biology ,Endoplasmic Reticulum ,protein interactions ,Biochemistry ,genomic ,Mice ,Molecular Basis of Cell and Developmental Biology ,Stress, Physiological ,Two-Hybrid System Techniques ,Heat shock protein ,genomics ,Animals ,Gene Knock-In Techniques ,RNA, Messenger ,protein interaction ,Molecular Biology ,Gene ,Cells, Cultured ,Heat-Shock Proteins ,Dominance (genetics) ,Neurons ,gene expression ,neurodegeneration ,Huntington's disease ,Endoplasmic reticulum ,Membrane Proteins ,RNA-Binding Proteins ,Cell Biology ,Molecular biology ,Corpus Striatum ,Mice, Mutant Strains ,Cell biology ,Mice, Inbred C57BL ,Disease Models, Animal ,Huntington Disease ,Membrane protein ,Unfolded protein response ,Cell Nucleolus - Abstract
The induction of Rrs1 expression is one of the earliest events detected in a presymptomatic knock-in mouse model of Huntington disease (HD). Rrs1 up-regulation fulfills the HD criteria of dominance, striatal specificity, and polyglutamine dependence. Here we show that mammalian Rrs1 is localized both in the nucleolus as well as in the endoplasmic reticulum (ER) of neurons. This dual localization is shared with its newly identified molecular partner 3D3/lyric. We then show that both genes are induced by ER stress in neurons. Interestingly, we demonstrate that ER stress is an early event in a presymptomatic HD mouse model that persists throughout the life span of the rodent. We further show that ER stress also occurs in postmortem brains of HD patients.
- Published
- 2009
- Full Text
- View/download PDF
29. Ser46 phosphorylation and prolyl-isomerase Pin1-mediated isomerization of p53 are key events in p53-dependent apoptosis induced by mutant huntingtin
- Author
-
Stefano Gustincich, Giannino Del Sal, Francesca Persichetti, Anna Comel, Fiamma Mantovani, Alice Grison, Elena Agostoni, Grison, A., Mantovani, Fiamma, Comel, Anna, Agostoni, E., Gustincich, S., Persichetti, F., and DEL SAL, Giannino
- Subjects
Huntingtin ,Apoptosis Inhibitor ,Parkinson's disease ,Nerve Tissue Proteins ,Apoptosis ,Biology ,Mice ,Isomerism ,neurodeneration ,Serine ,Huntingtin Protein ,Prolyl isomerase ,medicine ,Animals ,Humans ,Phosphorylation ,Protein kinase A ,Cell proliferation ,Peptidylprolyl isomerase ,prolyl-isomerization ,Multidisciplinary ,Huntington disease ,Neurodegeneration ,Nuclear Proteins ,Biological Sciences ,Peptidylprolyl Isomerase ,medicine.disease ,Molecular biology ,Cell biology ,Oxidative Stress ,Mutation ,PIN1 ,Tumor Suppressor Protein p53 ,DNA Damage - Abstract
Huntington disease (HD) is a neurodegenerative disorder caused by a CAG repeat expansion in the gene coding for huntingtin protein. Several mechanisms have been proposed by which mutant huntingtin (mHtt) may trigger striatal neurodegeneration, including mitochondrial dysfunction, oxidative stress, and apoptosis. Furthermore, mHtt induces DNA damage and activates a stress response. In this context, p53 plays a crucial role in mediating mHtt toxic effects. Here we have dissected the pathway of p53 activation by mHtt in human neuronal cells and in HD mice, with the aim of highlighting critical nodes that may be pharmacologically manipulated for therapeutic intervention. We demonstrate that expression of mHtt causes increased phosphorylation of p53 on Ser46, leading to its interaction with phosphorylation-dependent prolyl isomerase Pin1 and consequent dissociation from the apoptosis inhibitor iASPP, thereby inducing the expression of apoptotic target genes. Inhibition of Ser46 phosphorylation by targeting homeodomain-interacting protein kinase 2 (HIPK2), PKCδ, or ataxia telangiectasia mutated kinase, as well as inhibition of the prolyl isomerase Pin1, prevents mHtt-dependent apoptosis of neuronal cells. These results provide a rationale for the use of small-molecule inhibitors of stress-responsive protein kinases and Pin1 as a potential therapeutic strategy for HD treatment.
- Published
- 2011
30. Giant chess game enhances spatial navigational skills in 6-years-old children: preliminary findings.
- Author
-
Bocchi A, Palmiero M, Persichetti F, Matteoli M, Guariglia C, and Piccardi L
- Subjects
- Child, Humans, Memory, Short-Term, Spatial Memory, Confusion, Space Perception, Spatial Navigation
- Abstract
The game of chess is a valuable extracurricular activity for children, with positive effects on their cognitive skills and academic achievements. We investigated the extent to which the Giant Chess Game (GCG) played on a giant chessboard enhances working memory in "navigational-vista" space and "reaching" space. We also assessed if the GCG enhances mental rotation skills. For 10 weeks, 15 children (GCG group) were involved in a giant chess class, while 15 gender and age-matched children were involved in standard didactics (control group-CG). Children were tested twice, before (T0) and after (T1) the GCG, by tasks aimed at measuring: visuo-spatial working memory (VSWM) in the navigational-vista space (Walking Corsi test); VSWM in the reaching space (Corsi Block-Tapping task); mental rotation (Rotating Flowers test). We found that the GCG group significantly improved its performance more than the CG in VSWM in both navigational-vista space and reaching space, as well as in mental rotation. Our results suggest that the GCG has positive effects on visuo-spatial abilities underlying topographical skills. Therefore, the training using GCG can help enhancing spatial ability and may have a role in contrasting the spreading of navigational deficits such as the Developmental Topographical Disorientation (DTD).
- Published
- 2024
- Full Text
- View/download PDF
31. LINE-1 regulates cortical development by acting as long non-coding RNAs.
- Author
-
Mangoni D, Simi A, Lau P, Armaos A, Ansaloni F, Codino A, Damiani D, Floreani L, Di Carlo V, Vozzi D, Persichetti F, Santoro C, Pandolfini L, Tartaglia GG, Sanges R, and Gustincich S
- Subjects
- Animals, Mice, Cell Differentiation, Chromatin genetics, Chromatin Assembly and Disassembly, Long Interspersed Nucleotide Elements genetics, RNA, Long Noncoding genetics
- Abstract
Long Interspersed Nuclear Elements-1s (L1s) are transposable elements that constitute most of the genome's transcriptional output yet have still largely unknown functions. Here we show that L1s are required for proper mouse brain corticogenesis operating as regulatory long non-coding RNAs. They contribute to the regulation of the balance between neuronal progenitors and differentiation, the migration of post-mitotic neurons and the proportions of different cell types. In cortical cultured neurons, L1 RNAs are mainly associated to chromatin and interact with the Polycomb Repressive Complex 2 (PRC2) protein subunits enhancer of Zeste homolog 2 (Ezh2) and suppressor of zeste 12 (Suz12). L1 RNA silencing influences PRC2's ability to bind a portion of its targets and the deposition of tri-methylated histone H3 (H3K27me3) marks. Our results position L1 RNAs as crucial signalling hubs for genome-wide chromatin remodelling, enabling the fine-tuning of gene expression during brain development and evolution., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
32. SINEUP non-coding RNA activity depends on specific N6-methyladenosine nucleotides.
- Author
-
Pierattini B, D'Agostino S, Bon C, Peruzzo O, Alendar A, Codino A, Ros G, Persichetti F, Sanges R, Carninci P, Santoro C, Espinoza S, Valentini P, Pandolfini L, and Gustincich S
- Abstract
SINEUPs are natural and synthetic antisense long non-coding RNAs (lncRNAs) selectively enhancing target mRNAs translation by increasing their association with polysomes. This activity requires two RNA domains: an embedded inverted SINEB2 element acting as effector domain, and an antisense region, the binding domain, conferring target selectivity. SINEUP technology presents several advantages to treat genetic (haploinsufficiencies) and complex diseases restoring the physiological activity of diseased genes and of compensatory pathways. To streamline these applications to the clinic, a better understanding of the mechanism of action is needed. Here we show that natural mouse SINEUP AS Uchl1 and synthetic human miniSINEUP-DJ-1 are N
6 -methyladenosine (m6 A) modified by METTL3 enzyme. Then, we map m6 A-modified sites along SINEUP sequence with Nanopore direct RNA sequencing and a reverse transcription assay. We report that m6 A removal from SINEUP RNA causes the depletion of endogenous target mRNA from actively translating polysomes, without altering SINEUP enrichment in ribosomal subunit-associated fractions. These results prove that SINEUP activity requires an m6 A-dependent step to enhance translation of target mRNAs, providing a new mechanism for m6 A translation regulation and strengthening our knowledge of SINEUP-specific mode of action. Altogether these new findings pave the way to a more effective therapeutic application of this well-defined class of lncRNAs., Competing Interests: S.G., C.S., and P.C. are co-founders of Transine Therapeutics., (© 2023 The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
33. Neuronal haemoglobin induces loss of dopaminergic neurons in mouse Substantia nigra, cognitive deficits and cleavage of endogenous α-synuclein.
- Author
-
Santulli C, Bon C, De Cecco E, Codrich M, Narkiewicz J, Parisse P, Perissinotto F, Santoro C, Persichetti F, Legname G, Espinoza S, and Gustincich S
- Subjects
- Mice, Animals, Dopaminergic Neurons metabolism, Substantia Nigra metabolism, Hemoglobins metabolism, Cognition, alpha-Synuclein genetics, alpha-Synuclein metabolism, Parkinson Disease metabolism
- Abstract
Parkinson's disease (PD) presents the selective loss of A9 dopaminergic (DA) neurons of Substantia Nigra pars compacta (SNpc) and the presence of intracellular aggregates called Lewy bodies. α-synuclein (α-syn) species truncated at the carboxy-terminal (C-terminal) accumulate in pathological inclusions and promote α-syn aggregation and toxicity. Haemoglobin (Hb) is the major oxygen carrier protein in erythrocytes. In addition, Hb is expressed in A9 DA neurons where it influences mitochondrial activity. Hb overexpression increases cells' vulnerability in a neurochemical model of PD in vitro and forms cytoplasmic and nucleolar aggregates upon short-term overexpression in mouse SNpc. In this study, α and β-globin chains were co-expressed in DA cells of SNpc in vivo upon stereotaxic injections of an Adeno-Associated Virus isotype 9 (AAV9) and in DA iMN9D cells in vitro. Long-term Hb over-expression in SNpc induced the loss of about 50% of DA neurons, mild motor impairments, and deficits in recognition and spatial working memory. Hb triggered the formation of endogenous α-syn C-terminal truncated species. Similar α-syn fragments were found in vitro in DA iMN9D cells over-expressing α and β- globins when treated with pre-formed α-syn fibrils. Our study positions Hb as a relevant player in PD pathogenesis for its ability to trigger DA cells' loss in vivo and the formation of C-terminal α-syn fragments., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
34. Specific transcriptional programs differentiate ICOS from CD28 costimulatory signaling in human Naïve CD4 + T cells.
- Author
-
Gigliotti CL, Boggio E, Favero F, Incarnato D, Santoro C, Oliviero S, Rojo JM, Zucchelli S, Persichetti F, Baldanzi G, Dianzani U, and Corà D
- Subjects
- Cholesterol metabolism, Glycosaminoglycans metabolism, Humans, Inducible T-Cell Co-Stimulator Protein metabolism, Receptors, Antigen, T-Cell metabolism, Transcription, Genetic, p38 Mitogen-Activated Protein Kinases metabolism, CD28 Antigens, CD4-Positive T-Lymphocytes
- Abstract
Costimulatory molecules of the CD28 family play a crucial role in the activation of immune responses in T lymphocytes, complementing and modulating signals originating from the T-cell receptor (TCR) complex. Although distinct functional roles have been demonstrated for each family member, the specific signaling pathways differentiating ICOS- from CD28-mediated costimulation during early T-cell activation are poorly characterized. In the present study, we have performed RNA-Seq-based global transcriptome profiling of anti-CD3-treated naïve CD4
+ T cells upon costimulation through either inducible costimulator (ICOS) or CD28, revealing a set of signaling pathways specifically associated with each signal. In particular, we show that CD3/ICOS costimulation plays a major role in pathways related to STAT3 function and osteoarthritis (OA), whereas the CD3/CD28 axis mainly regulates p38 MAPK signaling. Furthermore, we report the activation of distinct immunometabolic pathways, with CD3/ICOS costimulation preferentially targeting glycosaminoglycans (GAGs) and CD3/CD28 regulating mitochondrial respiratory chain and cholesterol biosynthesis. These data suggest that ICOS and CD28 costimulatory signals play distinct roles during the activation of naïve T cells by modulating distinct sets of immunological and immunometabolic genes., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Gigliotti, Boggio, Favero, Incarnato, Santoro, Oliviero, Rojo, Zucchelli, Persichetti, Baldanzi, Dianzani and Corà.)- Published
- 2022
- Full Text
- View/download PDF
35. The E3 Ubiquitin Ligase TRAF6 Interacts with the Cellular Prion Protein and Modulates Its Solubility and Recruitment to Cytoplasmic p62/SQSTM1-Positive Aggresome-Like Structures.
- Author
-
Masperone L, Codrich M, Persichetti F, Gustincich S, Zucchelli S, and Legname G
- Subjects
- Animals, Mice, Prion Proteins metabolism, Protein Binding, Sequestosome-1 Protein metabolism, Solubility, TNF Receptor-Associated Factor 6 metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
The cellular prion protein (PrP
C ) is a ubiquitous glycoprotein highly expressed in the brain where it is involved in neurite outgrowth, copper homeostasis, NMDA receptor regulation, cell adhesion, and cell signaling. Conformational conversion of PrPC into its insoluble and aggregation-prone scrapie form (PrPSc ) is the trigger for several rare devastating neurodegenerative disorders, collectively referred to as prion diseases. Recent work indicates that the ubiquitin-proteasome system is involved in quality control of PrPC . To better dissect the role of ubiquitination in PrPC physiology, we focused on the E3 RING ubiquitin ligase tumor necrosis factor receptor (TNFR)-associated factor 6 (TRAF6). Here, we report that PrPC interacts with TRAF6 both in vitro, in cells, and in vivo, in the mouse brain. Transient overexpression of TRAF6 indirectly modulates PrPC ubiquitination and triggers redistribution of PrPC into the insoluble fraction. Importantly, in the presence of wild-type TRAF6, but not a mutant lacking E3 ligase activity, PrPC accumulates into cytoplasmic aggresome-like inclusions containing TRAF6 and p62/SQSTM1. Our results suggest that TRAF6 ligase activity could exert a role in the regulation of PrPC redistribution in cells under physiological conditions. This novel interaction may uncover possible mechanisms of cell clearance/reorganization in prion diseases., (© 2021. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)- Published
- 2022
- Full Text
- View/download PDF
36. Analysis of LINE1 Retrotransposons in Huntington's Disease.
- Author
-
Floreani L, Ansaloni F, Mangoni D, Agostoni E, Sanges R, Persichetti F, and Gustincich S
- Abstract
Transposable elements (TEs) are mobile genetic elements that made up about half the human genome. Among them, the autonomous non-LTR retrotransposon long interspersed nuclear element-1 (L1) is the only currently active TE in mammals and covers about 17% of the mammalian genome. L1s exert their function as structural elements in the genome, as transcribed RNAs to influence chromatin structure and as retrotransposed elements to shape genomic variation in somatic cells. L1s activity has been shown altered in several diseases of the nervous system. Huntington disease (HD) is a dominantly inherited neurodegenerative disorder caused by an expansion of a CAG repeat in the HTT gene which leads to a gradual loss of neurons most prominently in the striatum and, to a lesser extent, in cortical brain regions. The length of the expanded CAG tract is related to age at disease onset, with longer repeats leading to earlier onset. Here we carried out bioinformatic analysis of public RNA-seq data of a panel of HD mouse models showing that a decrease of L1 RNA expression recapitulates two hallmarks of the disease: it correlates to CAG repeat length and it occurs in the striatum, the site of neurodegeneration. Results were then experimentally validated in Htt
Q 111 knock-in mice. The expression of L1-encoded proteins was independent from L1 RNA levels and differentially regulated in time and tissues. The pattern of expression L1 RNAs in human HD post-mortem brains showed similarity to mouse models of the disease. This work suggests the need for further study of L1s in HD and adds support to the current hypothesis that dysregulation of TEs may be involved in neurodegenerative diseases., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Floreani, Ansaloni, Mangoni, Agostoni, Sanges, Persichetti and Gustincich.)- Published
- 2022
- Full Text
- View/download PDF
37. Transposable element activation promotes neurodegeneration in a Drosophila model of Huntington's disease.
- Author
-
Casale AM, Liguori F, Ansaloni F, Cappucci U, Finaurini S, Spirito G, Persichetti F, Sanges R, Gustincich S, and Piacentini L
- Abstract
Huntington's disease (HD) is an autosomal dominant disorder with progressive motor dysfunction and cognitive decline. The disease is caused by a CAG repeat expansion in the IT15 gene, which elongates a polyglutamine stretch of the HD protein, Huntingtin. No therapeutic treatments are available, and new pharmacological targets are needed. Retrotransposons are transposable elements (TEs) that represent 40% and 30% of the human and Drosophila genomes and replicate through an RNA intermediate. Mounting evidence suggests that mammalian TEs are active during neurogenesis and may be involved in diseases of the nervous system. Here we show that TE expression and mobilization are increased in a Drosophila melanogaster HD model. By inhibiting TE mobilization with Reverse Transcriptase inhibitors, polyQ-dependent eye neurodegeneration and genome instability in larval brains are rescued and fly lifespan is increased. These results suggest that TE activation may be involved in polyQ-induced neurotoxicity and a potential pharmacological target., Competing Interests: The authors declare no competing interests., (© 2021 The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
38. SINEUPs: a novel toolbox for RNA therapeutics.
- Author
-
Espinoza S, Bon C, Valentini P, Pierattini B, Matey AT, Damiani D, Pulcrano S, Sanges R, Persichetti F, Takahashi H, Carninci P, Santoro C, Cotella D, and Gustincich S
- Subjects
- Animals, Mice, Proteins metabolism, RNA, Antisense genetics, RNA, Antisense metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Protein Biosynthesis, RNA, Long Noncoding chemistry
- Abstract
RNA molecules have emerged as a new class of promising therapeutics to expand the range of druggable targets in the genome. In addition to 'canonical' protein-coding mRNAs, the emerging richness of sense and antisense long non-coding RNAs (lncRNAs) provides a new reservoir of molecular tools for RNA-based drugs. LncRNAs are composed of modular structural domains with specific activities involving the recruitment of protein cofactors or directly interacting with nucleic acids. A single therapeutic RNA transcript can then be assembled combining domains with defined secondary structures and functions, and antisense sequences specific for the RNA/DNA target of interest. As the first representative molecules of this new pharmacology, we have identified SINEUPs, a new functional class of natural antisense lncRNAs that increase the translation of partially overlapping mRNAs. Their activity is based on the combination of two domains: an embedded mouse inverted SINEB2 element that enhances mRNA translation (effector domain) and an overlapping antisense region that provides specificity for the target sense transcript (binding domain). By genetic engineering, synthetic SINEUPs can potentially target any mRNA of interest increasing translation and therefore the endogenous level of the encoded protein. In this review, we describe the state-of-the-art knowledge of SINEUPs and discuss recent publications showing their potential application in diseases where a physiological increase of endogenous protein expression can be therapeutic., (© 2021 The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
39. A human minisatellite hosts an alternative transcription start site for NPRL3 driving its expression in a repeat number-dependent manner.
- Author
-
Bertuzzi M, Tang D, Calligaris R, Vlachouli C, Finaurini S, Sanges R, Goldwurm S, Catalan M, Antonutti L, Manganotti P, Pizzolato G, Pezzoli G, Persichetti F, Carninci P, and Gustincich S
- Subjects
- Cell Line, GTPase-Activating Proteins genetics, Genomics methods, Genotype, Humans, Introns, Multigene Family, Polymorphism, Genetic, RNA Caps, RNA Interference, RNA, Small Interfering, GTPase-Activating Proteins metabolism, Gene Expression Regulation, Minisatellite Repeats, Transcription Initiation Site
- Abstract
Minisatellites, also called variable number of tandem repeats (VNTRs), are a class of repetitive elements that may affect gene expression at multiple levels and have been correlated to disease. Their identification and role as expression quantitative trait loci (eQTL) have been limited by their absence in comparative genomic hybridization and single nucleotide polymorphisms arrays. By taking advantage of cap analysis of gene expression (CAGE), we describe a new example of a minisatellite hosting a transcription start site (TSS) which expression is dependent on the repeat number. It is located in the third intron of the gene nitrogen permease regulator like protein 3 (NPRL3). NPRL3 is a component of the GAP activity toward rags 1 protein complex that inhibits mammalian target of rapamycin complex 1 (mTORC1) activity and it is found mutated in familial focal cortical dysplasia and familial focal epilepsy. CAGE tags represent an alternative TSS identifying TAGNPRL3 messenger RNAs (mRNAs). TAGNPRL3 is expressed in red blood cells both at mRNA and protein levels, it interacts with its protein partner NPRL2 and its overexpression inhibits cell proliferation. This study provides an example of a minisatellite that is both a TSS and an eQTL as well as identifies a new VNTR that may modify mTORC1 activity., (© 2020 The Authors. Human Mutation published by Wiley Periodicals, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
40. The RNA-binding protein ILF3 binds to transposable element sequences in SINEUP lncRNAs.
- Author
-
Fasolo F, Patrucco L, Volpe M, Bon C, Peano C, Mignone F, Carninci P, Persichetti F, Santoro C, Zucchelli S, Sblattero D, Sanges R, Cotella D, and Gustincich S
- Subjects
- Animals, Computational Biology, High-Throughput Screening Assays, Humans, Mice, Nuclear Factor 90 Proteins genetics, Protein Biosynthesis, Protein Interaction Domains and Motifs, RNA, Long Noncoding genetics, RNA, Messenger genetics, Ubiquitin Thiolesterase genetics, DNA Transposable Elements, Nuclear Factor 90 Proteins metabolism, RNA, Antisense genetics, RNA, Long Noncoding metabolism, RNA, Messenger metabolism, Ubiquitin Thiolesterase metabolism
- Abstract
Transposable elements (TEs) compose about half of the mammalian genome and, as embedded sequences, up to 40% of long noncoding RNA (lncRNA) transcripts. Embedded TEs may represent functional domains within lncRNAs, providing a structured RNA platform for protein interaction. Here we show the interactome profile of the mouse inverted short interspersed nuclear element (SINE) of subfamily B2 (invSINEB2) alone and embedded in antisense (AS) ubiquitin C-terminal hydrolase L1 (Uchl1), an lncRNA that is AS to Uchl1 gene. AS Uchl1 is the representative member of a functional class of AS lncRNAs, named SINEUPs, in which the invSINEB2 acts as effector domain (ED)-enhancing translation of sense protein-coding mRNAs. By using RNA-interacting domainome technology, we identify the IL enhancer-binding factor 3 (ILF3) as a protein partner of AS Uchl1 RNA. We determine that this interaction is mediated by the RNA-binding motif 2 of ILF3 and the invSINEB2. Furthermore, we show that ILF3 is able to bind a free right Arthrobacter luteus (Alu) monomer sequence, the embedded TE acting as ED in human SINEUPs. Bioinformatic analysis of Encyclopedia of DNA Elements-enhanced cross-linking immunoprecipitation data reveals that ILF3 binds transcribed human SINE sequences at transcriptome-wide levels. We then demonstrate that the embedded TEs modulate AS Uchl1 RNA nuclear localization to an extent moderately influenced by ILF3. This work unveils the existence of a specific interaction between embedded TEs and an RNA-binding protein, strengthening the model of TEs as functional modules in lncRNAs.-Fasolo, F., Patrucco, L., Volpe, M., Bon, C., Peano, C., Mignone, F., Carninci, P., Persichetti, F., Santoro, C., Zucchelli, S., Sblattero, D., Sanges, R., Cotella, D., Gustincich, S. The RNA-binding protein ILF3 binds to transposable element sequences in SINEUP lncRNAs.
- Published
- 2019
- Full Text
- View/download PDF
41. SINEUP non-coding RNAs rescue defective frataxin expression and activity in a cellular model of Friedreich's Ataxia.
- Author
-
Bon C, Luffarelli R, Russo R, Fortuni S, Pierattini B, Santulli C, Fimiani C, Persichetti F, Cotella D, Mallamaci A, Santoro C, Carninci P, Espinoza S, Testi R, Zucchelli S, Condò I, and Gustincich S
- Subjects
- Aconitate Hydratase metabolism, Cell Line, Fibroblasts metabolism, Humans, Lymphocytes metabolism, Phenotype, RNA, Messenger genetics, RNA, Messenger metabolism, RNA, Untranslated genetics, Frataxin, Friedreich Ataxia genetics, Gene Expression Regulation, Iron-Binding Proteins genetics, Models, Biological, RNA, Untranslated metabolism
- Abstract
Friedreich's ataxia (FRDA) is an untreatable disorder with neuro- and cardio-degenerative progression. This monogenic disease is caused by the hyper-expansion of naturally occurring GAA repeats in the first intron of the FXN gene, encoding for frataxin, a protein implicated in the biogenesis of iron-sulfur clusters. As the genetic defect interferes with FXN transcription, FRDA patients express a normal frataxin protein but at insufficient levels. Thus, current therapeutic strategies are mostly aimed to restore physiological FXN expression. We have previously described SINEUPs, natural and synthetic antisense long non-coding RNAs, which promote translation of partially overlapping mRNAs through the activity of an embedded SINEB2 domain. Here, by in vitro screening, we have identified a number of SINEUPs targeting human FXN mRNA and capable to up-regulate frataxin protein to physiological amounts acting at the post-transcriptional level. Furthermore, FXN-specific SINEUPs promote the recovery of disease-associated mitochondrial aconitase defects in FRDA-derived cells. In summary, we provide evidence that SINEUPs may be the first gene-specific therapeutic approach to activate FXN translation in FRDA and, more broadly, a novel scalable platform to develop new RNA-based therapies for haploinsufficient diseases., (© The Author(s) 2019. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2019
- Full Text
- View/download PDF
42. Antisense Transcription in Loci Associated to Hereditary Neurodegenerative Diseases.
- Author
-
Zucchelli S, Fedele S, Vatta P, Calligaris R, Heutink P, Rizzu P, Itoh M, Persichetti F, Santoro C, Kawaji H, Lassmann T, Hayashizaki Y, Carninci P, Forrest ARR, and Gustincich S
- Subjects
- Cell Line, Tumor, Gene Expression Regulation, Genetic Pleiotropy, Humans, Molecular Sequence Annotation, RNA, Long Noncoding genetics, RNA, Long Noncoding metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Genetic Loci, Genetic Predisposition to Disease, Neurodegenerative Diseases genetics, RNA, Antisense genetics, Transcription, Genetic
- Abstract
Natural antisense transcripts are common features of mammalian genes providing additional regulatory layers of gene expression. A comprehensive description of antisense transcription in loci associated to familial neurodegenerative diseases may identify key players in gene regulation and provide tools for manipulating gene expression. We take advantage of the FANTOM5 sequencing datasets that represent the largest collection to date of genome-wide promoter usage in almost 2000 human samples. Transcription start sites (TSSs) are mapped at high resolution by the use of a modified protocol of cap analysis of gene expression (CAGE) for high-throughput single molecule next-generation sequencing with Helicos (hCAGE). Here we present the analysis of antisense transcription at 17 loci associated to hereditary Alzheimer's disease, Frontotemporal Dementia, Parkinson's disease, Amyotrophic Lateral Sclerosis, and Huntington's disease. We focused our analysis on libraries derived from brain tissues and primary cells. We also screened libraries from total blood and blood cell populations in the quest for peripheral biomarkers of neurodegenerative diseases. We identified 63 robust promoters in antisense orientation to genes associated to familial neurodegeneration. When applying a less stringent cutoff, this number increases to over 400. A subset of these promoters represents alternative TSSs for 24 FANTOM5 annotated long noncoding RNA (lncRNA) genes, in antisense orientation to 13 of the loci analyzed here, while the remaining contribute to the expression of additional transcript variants. Intersection with GWAS studies, sample ontology, and dynamic expression reveals association to specific genetic traits as well as cell and tissue types, not limited to neurodegenerative diseases. Antisense transcription was validated for a subset of genes, including those encoding for Microtubule-Associated Protein Tau, α-synuclein, Parkinsonism-associated deglycase DJ-1, and Leucin-Rich Repeat Kinase 2. This work provides evidence for the existence of additional regulatory mechanisms of the expression of neurodegenerative disease-causing genes by previously not-annotated and/or not-validated antisense long noncoding RNAs.
- Published
- 2019
- Full Text
- View/download PDF
43. Huntingtin polyQ Mutation Impairs the 17β-Estradiol/Neuroglobin Pathway Devoted to Neuron Survival.
- Author
-
Nuzzo MT, Fiocchetti M, Totta P, Melone MAB, Cardinale A, Fusco FR, Gustincich S, Persichetti F, Ascenzi P, and Marino M
- Subjects
- Animals, Cell Line, Tumor, Cell Survival drug effects, Corpus Striatum cytology, Corpus Striatum drug effects, Corpus Striatum metabolism, Hippocampus cytology, Hippocampus drug effects, Hippocampus metabolism, Mice, Mice, Transgenic, Neuroglobin, Neurons cytology, Neurons drug effects, Neuroprotection drug effects, Neuroprotection genetics, Signal Transduction drug effects, Up-Regulation drug effects, Cell Survival genetics, Estradiol pharmacology, Globins metabolism, Huntingtin Protein genetics, Mutation, Nerve Tissue Proteins metabolism, Neurons metabolism, Peptides genetics, Signal Transduction genetics
- Abstract
Among several mechanisms underlying the well-known trophic and protective effects of 17β-estradiol (E2) in the brain, we recently reported that E2 induces the up-regulation of two anti-apoptotic and neuroprotectant proteins: huntingtin (HTT) and neuroglobin (NGB). Here, we investigate the role of this up-regulation. The obtained results indicate that E2 promotes NGB-HTT association, induces the localization of the complex at the mitochondria, and protects SK-N-BE neuroblastoma cells and murine striatal cells, which express wild-type HTT (i.e., polyQ
7 ), against H2 O2 -induced apoptosis. All E2 effects were completely abolished in HTT-knocked out SK-N-BE cells and in striatal neurons expressing the mutated form of HTT (mHTT; i.e., polyQ111 ) typical of Huntington's disease (HD). As a whole, these data provide a new function of wild-type HTT which drives E2-induced NGB in mitochondria modulating NGB anti-apoptotic activity. This new function is lost by HTT polyQ pathological expansion. These data evidence the existence of a novel E2/HTT/NGB neuroprotective axis that may play a relevant role in the development of HD therapeutics.- Published
- 2017
- Full Text
- View/download PDF
44. Neuronal hemoglobin affects dopaminergic cells' response to stress.
- Author
-
Codrich M, Bertuzzi M, Russo R, Francescatto M, Espinoza S, Zentilin L, Giacca M, Cesselli D, Beltrami AP, Ascenzi P, Zucchelli S, Persichetti F, Leanza G, and Gustincich S
- Subjects
- 1-Methyl-4-phenylpyridinium toxicity, Animals, Autophagy genetics, Brain metabolism, Brain pathology, Dopaminergic Neurons pathology, Epigenesis, Genetic genetics, Gene Expression drug effects, Hemoglobins biosynthesis, Hemoglobins metabolism, Humans, Mice, Parkinson Disease metabolism, Parkinson Disease pathology, Parkinson Disease, Secondary pathology, Rotenone toxicity, Substantia Nigra metabolism, Substantia Nigra pathology, Dopaminergic Neurons metabolism, Hemoglobins genetics, Parkinson Disease genetics, Parkinson Disease, Secondary genetics
- Abstract
Hemoglobin (Hb) is the major protein in erythrocytes and carries oxygen (O
2 ) throughout the body. Recently, Hb has been found synthesized in atypical sites, including the brain. Hb is highly expressed in A9 dopaminergic (DA) neurons of the substantia nigra (SN), whose selective degeneration leads to Parkinson's disease (PD). Here we show that Hb confers DA cells' susceptibility to 1-methyl-4-phenylpyridinium (MPP+ ) and rotenone, neurochemical cellular models of PD. The toxic property of Hb does not depend on O2 binding and is associated with insoluble aggregate formation in the nucleolus. Neurochemical stress induces epigenetic modifications, nucleolar alterations and autophagy inhibition that depend on Hb expression. When adeno-associated viruses carrying α- and β-chains of Hb are stereotaxically injected into mouse SN, Hb forms aggregates and causes motor learning impairment. These results position Hb as a potential player in DA cells' homeostasis and dysfunction in PD.- Published
- 2017
- Full Text
- View/download PDF
45. Engineering Translation in Mammalian Cell Factories to Increase Protein Yield: The Unexpected Use of Long Non-Coding SINEUP RNAs.
- Author
-
Zucchelli S, Patrucco L, Persichetti F, Gustincich S, and Cotella D
- Abstract
Mammalian cells are an indispensable tool for the production of recombinant proteins in contexts where function depends on post-translational modifications. Among them, Chinese Hamster Ovary (CHO) cells are the primary factories for the production of therapeutic proteins, including monoclonal antibodies (MAbs). To improve expression and stability, several methodologies have been adopted, including methods based on media formulation, selective pressure and cell- or vector engineering. This review presents current approaches aimed at improving mammalian cell factories that are based on the enhancement of translation. Among well-established techniques (codon optimization and improvement of mRNA secondary structure), we describe SINEUPs, a family of antisense long non-coding RNAs that are able to increase translation of partially overlapping protein-coding mRNAs. By exploiting their modular structure, SINEUP molecules can be designed to target virtually any mRNA of interest, and thus to increase the production of secreted proteins. Thus, synthetic SINEUPs represent a new versatile tool to improve the production of secreted proteins in biomanufacturing processes.
- Published
- 2016
- Full Text
- View/download PDF
46. Effects of Pin1 Loss in Hdh(Q111) Knock-in Mice.
- Author
-
Agostoni E, Michelazzi S, Maurutto M, Carnemolla A, Ciani Y, Vatta P, Roncaglia P, Zucchelli S, Leanza G, Mantovani F, Gustincich S, Santoro C, Piazza S, Del Sal G, and Persichetti F
- Abstract
Huntington's disease (HD) is a fatal, dominantly inherited, neurodegenerative disorder due to a pathological expansion of the CAG repeat in the coding region of the HTT gene. In the quest for understanding the molecular basis of neurodegeneration, we have previously demonstrated that the prolyl isomerase Pin1 plays a crucial role in mediating p53-dependent apoptosis triggered by mutant huntingtin (mHtt) in vitro. To assess the effects of the lack of Pin1 in vivo, we have bred Pin1 knock-out mice with Hdh(Q111) knock-in mice, a genetically precise model of HD. We show that Pin1 genetic ablation modifies a portion of Hdh(Q111) phenotypes in a time-dependent fashion. As an early event, Pin1 activity reduces the DNA damage response (DDR). In midlife mice, by taking advantage of next-generation sequencing technology, we show that Pin1 activity modulates a portion of the alterations triggered by mHtt, extending the role of Pin1 to two additional Hdh(Q111) phenotypes: the unbalance in the "synthesis/concentration of hormones", as well as the alteration of "Wnt/β-catenin signaling". In aging animals, Pin1 significantly increases the number of mHtt-positive nuclear inclusions while it reduces gliosis. In summary, this work provides further support for a role of Pin1 in HD pathogenesis.
- Published
- 2016
- Full Text
- View/download PDF
47. SINEUPs are modular antisense long non-coding RNAs that increase synthesis of target proteins in cells.
- Author
-
Zucchelli S, Fasolo F, Russo R, Cimatti L, Patrucco L, Takahashi H, Jones MH, Santoro C, Sblattero D, Cotella D, Persichetti F, Carninci P, and Gustincich S
- Abstract
Despite recent efforts in discovering novel long non-coding RNAs (lncRNAs) and unveiling their functions in a wide range of biological processes their applications as biotechnological or therapeutic tools are still at their infancy. We have recently shown that AS Uchl1, a natural lncRNA antisense to the Parkinson's disease-associated gene Ubiquitin carboxyl-terminal esterase L1 (Uchl1), is able to increase UchL1 protein synthesis at post-transcriptional level. Its activity requires two RNA elements: an embedded inverted SINEB2 sequence to increase translation and the overlapping region to target its sense mRNA. This functional organization is shared with several mouse lncRNAs antisense to protein coding genes. The potential use of AS Uchl1-derived lncRNAs as enhancers of target mRNA translation remains unexplored. Here we define AS Uchl1 as the representative member of a new functional class of natural and synthetic antisense lncRNAs that activate translation. We named this class of RNAs SINEUPs for their requirement of the inverted SINEB2 sequence to UP-regulate translation in a gene-specific manner. The overlapping region is indicated as the Binding Doman (BD) while the embedded inverted SINEB2 element is the Effector Domain (ED). By swapping BD, synthetic SINEUPs are designed targeting mRNAs of interest. SINEUPs function in an array of cell lines and can be efficiently directed toward N-terminally tagged proteins. Their biological activity is retained in a miniaturized version within the range of small RNAs length. Its modular structure was exploited to successfully design synthetic SINEUPs targeting endogenous Parkinson's disease-associated DJ-1 and proved to be active in different neuronal cell lines. In summary, SINEUPs represent the first scalable tool to increase synthesis of proteins of interest. We propose SINEUPs as reagents for molecular biology experiments, in protein manufacturing as well as in therapy of haploinsufficiencies.
- Published
- 2015
- Full Text
- View/download PDF
48. Expression analysis of the long non-coding RNA antisense to Uchl1 (AS Uchl1) during dopaminergic cells' differentiation in vitro and in neurochemical models of Parkinson's disease.
- Author
-
Carrieri C, Forrest AR, Santoro C, Persichetti F, Carninci P, Zucchelli S, and Gustincich S
- Abstract
Antisense (AS) transcripts are RNA molecules that are transcribed from the opposite strand to sense (S) genes forming S/AS pairs. The most prominent configuration is when a lncRNA is antisense to a protein coding gene. Increasing evidences prove that antisense transcription may control sense gene expression acting at distinct regulatory levels. However, its contribution to brain function and neurodegenerative diseases remains unclear. We have recently identified AS Uchl1 as an antisense to the mouse Ubiquitin carboxy-terminal hydrolase L1 (Uchl1) gene (AS Uchl1), the synthenic locus of UCHL1/PARK5. This is mutated in rare cases of early-onset familial Parkinson's Disease (PD) and loss of UCHL1 activity has been reported in many neurodegenerative diseases. Importantly, manipulation of UchL1 expression has been proposed as tool for therapeutic intervention. AS Uchl1 induces UchL1 expression by increasing its translation. It is the representative member of SINEUPs (SINEB2 sequence to UP-regulate translation), a new functional class of natural antisense lncRNAs that activate translation of their sense genes. Here we take advantage of FANTOM5 dataset to identify the transcription start sites associated to S/AS pair at Uchl1 locus. We show that AS Uchl1 expression is under the regulation of Nurr1, a major transcription factor involved in dopaminergic cells' differentiation and maintenance. Furthermore, AS Uch1 RNA levels are strongly down-regulated in neurochemical models of PD in vitro and in vivo. This work positions AS Uchl1 RNA as a component of Nurr1-dependent gene network and target of cellular stress extending our understanding on the role of antisense transcription in the brain.
- Published
- 2015
- Full Text
- View/download PDF
49. An Air-Well sparging minifermenter system for high-throughput protein production.
- Author
-
Deantonio C, Sedini V, Cesaro P, Quasso F, Cotella D, Persichetti F, Santoro C, and Sblattero D
- Subjects
- Escherichia coli growth & development, Escherichia coli metabolism, Protein Array Analysis, Air, Bioreactors microbiology, Biotechnology instrumentation, Biotechnology methods, High-Throughput Screening Assays methods, Recombinant Proteins biosynthesis
- Abstract
Background: Over the last few years High-Throughput Protein Production (HTPP) has played a crucial role for functional proteomics. High-quality, high yield and fast recombinant protein production are critical for new HTPP technologies. Escherichia coli is usually the expression system of choice in protein production thanks to its fast growth, ease of handling and high yields of protein produced. Even though shake-flask cultures are widely used, there is an increasing need for easy to handle, lab scale, high throughput systems., Results: In this article we described a novel minifermenter system suitable for HTPP. The Air-Well minifermenter system is made by a homogeneous air sparging device that includes an air diffusion system, and a stainless steel 96 needle plate integrated with a 96 deep well plate where cultures take place. This system provides aeration to achieve higher optical density growth compared to classical shaking growth without the decrease in pH value and bacterial viability. Moreover the yield of recombinant protein is up to 3-fold higher with a considerable improvement in the amount of full length proteins., Conclusions: High throughput production of hundreds of proteins in parallel can be obtained sparging air in a continuous and controlled manner. The system used is modular and can be easily modified and scaled up to meet the demands for HTPP.
- Published
- 2014
- Full Text
- View/download PDF
50. Mesencephalic dopaminergic neurons express a repertoire of olfactory receptors and respond to odorant-like molecules.
- Author
-
Grison A, Zucchelli S, Urzì A, Zamparo I, Lazarevic D, Pascarella G, Roncaglia P, Giorgetti A, Garcia-Esparcia P, Vlachouli C, Simone R, Persichetti F, Forrest AR, Hayashizaki Y, Carloni P, Ferrer I, Lodovichi C, Plessy C, Carninci P, and Gustincich S
- Subjects
- Animals, Cell Line, Cluster Analysis, Dopaminergic Neurons drug effects, Female, Gene Expression Profiling, Humans, Mice, Organ Specificity genetics, Parkinson Disease genetics, Parkinson Disease metabolism, Receptors, Odorant metabolism, Recombinant Proteins, Substantia Nigra metabolism, Transcription, Genetic, Dopaminergic Neurons metabolism, Gene Expression Regulation drug effects, Mesencephalon cytology, Mesencephalon metabolism, Odorants, Receptors, Odorant genetics
- Abstract
Background: The mesencephalic dopaminergic (mDA) cell system is composed of two major groups of projecting cells in the Substantia Nigra (SN) (A9 neurons) and the Ventral Tegmental Area (VTA) (A10 cells). Selective degeneration of A9 neurons occurs in Parkinson's disease (PD) while abnormal function of A10 cells has been linked to schizophrenia, attention deficit and addiction. The molecular basis that underlies selective vulnerability of A9 and A10 neurons is presently unknown., Results: By taking advantage of transgenic labeling, laser capture microdissection coupled to nano Cap-Analysis of Gene Expression (nanoCAGE) technology on isolated A9 and A10 cells, we found that a subset of Olfactory Receptors (OR)s is expressed in mDA neurons. Gene expression analysis was integrated with the FANTOM5 Helicos CAGE sequencing datasets, showing the presence of these ORs in selected tissues and brain areas outside of the olfactory epithelium. OR expression in the mesencephalon was validated by RT-PCR and in situ hybridization. By screening 16 potential ligands on 5 mDA ORs recombinantly expressed in an heterologous in vitro system, we identified carvone enantiomers as agonists at Olfr287 and able to evoke an intracellular Ca2+ increase in solitary mDA neurons. ORs were found expressed in human SN and down-regulated in PD post mortem brains., Conclusions: Our study indicates that mDA neurons express ORs and respond to odor-like molecules providing new opportunities for pharmacological intervention in disease.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.