49 results on '"Samuele Gherardi"'
Search Results
2. The DMD locus harbours multiple long non-coding RNAs which orchestrate and control transcription of muscle dystrophin mRNA isoforms.
- Author
-
Matteo Bovolenta, Daniela Erriquez, Emanuele Valli, Simona Brioschi, Chiara Scotton, Marcella Neri, Maria Sofia Falzarano, Samuele Gherardi, Marina Fabris, Paola Rimessi, Francesca Gualandi, Giovanni Perini, and Alessandra Ferlini
- Subjects
Medicine ,Science - Abstract
The 2.2 Mb long dystrophin (DMD) gene, the largest gene in the human genome, corresponds to roughly 0.1% of the entire human DNA sequence. Mutations in this gene cause Duchenne muscular dystrophy and other milder X-linked, recessive dystrophinopathies. Using a custom-made tiling array, specifically designed for the DMD locus, we identified a variety of novel long non-coding RNAs (lncRNAs), both sense and antisense oriented, whose expression profiles mirror that of DMD gene. Importantly, these transcripts are intronic in origin and specifically localized to the nucleus and are transcribed contextually with dystrophin isoforms or primed by MyoD-induced myogenic differentiation. Furthermore, their forced ectopic expression in both human muscle and neuronal cells causes a specific and negative regulation of endogenous dystrophin full length isoforms and significantly down-regulate the activity of a luciferase reporter construct carrying the minimal promoter regions of the muscle dystrophin isoform. Consistent with this apparently repressive role, we found that, in muscle samples of dystrophinopathic female carriers, lncRNAs expression levels inversely correlate with those of muscle full length DMD isoforms. Overall these findings unveil an unprecedented complexity of the transcriptional pattern of the DMD locus and reveal that DMD lncRNAs may contribute to the orchestration and homeostasis of the muscle dystrophin expression pattern by either selective targeting and down-modulating the dystrophin promoter transcriptional activity.
- Published
- 2012
- Full Text
- View/download PDF
3. SIRT1 promotes N-Myc oncogenesis through a positive feedback loop involving the effects of MKP3 and ERK on N-Myc protein stability.
- Author
-
Glenn M Marshall, Pei Y Liu, Samuele Gherardi, Christopher J Scarlett, Antonio Bedalov, Ning Xu, Nuncio Iraci, Emanuele Valli, Dora Ling, Wayne Thomas, Margo van Bekkum, Eric Sekyere, Kacper Jankowski, Toby Trahair, Karen L Mackenzie, Michelle Haber, Murray D Norris, Andrew V Biankin, Giovanni Perini, and Tao Liu
- Subjects
Genetics ,QH426-470 - Abstract
The N-Myc oncoprotein is a critical factor in neuroblastoma tumorigenesis which requires additional mechanisms converting a low-level to a high-level N-Myc expression. N-Myc protein is stabilized when phosphorylated at Serine 62 by phosphorylated ERK protein. Here we describe a novel positive feedback loop whereby N-Myc directly induced the transcription of the class III histone deacetylase SIRT1, which in turn increased N-Myc protein stability. SIRT1 binds to Myc Box I domain of N-Myc protein to form a novel transcriptional repressor complex at gene promoter of mitogen-activated protein kinase phosphatase 3 (MKP3), leading to transcriptional repression of MKP3, ERK protein phosphorylation, N-Myc protein phosphorylation at Serine 62, and N-Myc protein stabilization. Importantly, SIRT1 was up-regulated, MKP3 down-regulated, in pre-cancerous cells, and preventative treatment with the SIRT1 inhibitor Cambinol reduced tumorigenesis in TH-MYCN transgenic mice. Our data demonstrate the important roles of SIRT1 in N-Myc oncogenesis and SIRT1 inhibitors in the prevention and therapy of N-Myc-induced neuroblastoma.
- Published
- 2011
- Full Text
- View/download PDF
4. Supplementary Figures 1-4, Tables 1-2 from c-MYC Oncoprotein Dictates Transcriptional Profiles of ATP-Binding Cassette Transporter Genes in Chronic Myelogenous Leukemia CD34+ Hematopoietic Progenitor Cells
- Author
-
Giovanni Perini, Giovanni Martinelli, Michele Baccarani, Murray D. Norris, Michelle Haber, Chiara Perrod, Roberto Bernardoni, Thea Kalebic, Emanuele Valli, Sandra Durante, Carolina Terragna, Samuele Gherardi, Daniel Diolaiti, Simona Soverini, Nunzio Iraci, and Antonio Porro
- Abstract
PDF file - 2651K
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Dataset 2 from WDR5 Supports an N-Myc Transcriptional Complex That Drives a Protumorigenic Gene Expression Signature in Neuroblastoma
- Author
-
Tao Liu, Antony Braithwaite, Glenn M. Marshall, Giovanni Perini, Stefan Hüttelmaier, Karen L. MacKenzie, Masoud Vedadi, Cheryl H. Arrowsmith, Peter J. Brown, Johannes H. Schulte, Ygal Haupt, Jason M. Shohet, Quan Zhao, Toby Trahair, Matthew Wong, Bernard Atmadibrata, Bing Liu, Pei Y. Liu, Andrew E. Tee, Rima Al-Awar, Jason W.H. Wong, Giorgio Milazzo, Rebecca C. Poulos, Samuele Gherardi, Daniel Carter, Jessica L. Bell, and Yuting Sun
- Abstract
List of N-Myc-binding gene promoters at which H3K4me3 was reduced by WDR5 siRNA.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Dataset 3 from WDR5 Supports an N-Myc Transcriptional Complex That Drives a Protumorigenic Gene Expression Signature in Neuroblastoma
- Author
-
Tao Liu, Antony Braithwaite, Glenn M. Marshall, Giovanni Perini, Stefan Hüttelmaier, Karen L. MacKenzie, Masoud Vedadi, Cheryl H. Arrowsmith, Peter J. Brown, Johannes H. Schulte, Ygal Haupt, Jason M. Shohet, Quan Zhao, Toby Trahair, Matthew Wong, Bernard Atmadibrata, Bing Liu, Pei Y. Liu, Andrew E. Tee, Rima Al-Awar, Jason W.H. Wong, Giorgio Milazzo, Rebecca C. Poulos, Samuele Gherardi, Daniel Carter, Jessica L. Bell, and Yuting Sun
- Abstract
List of N-Myc non-binding gene promoters at which H3K4me3 was reduced by WDR5 siRNA.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figures 1-5 from p53 Is a Direct Transcriptional Target of MYCN in Neuroblastoma
- Author
-
Deborah A. Tweddle, John Lunec, Giovanni Perini, Katrina M. Wood, Laura D. Gamble, Samuele Gherardi, Nunzio Iraci, and Lindi Chen
- Abstract
Supplementary Figures 1-5 from p53 Is a Direct Transcriptional Target of MYCN in Neuroblastoma
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Dataset 4 from WDR5 Supports an N-Myc Transcriptional Complex That Drives a Protumorigenic Gene Expression Signature in Neuroblastoma
- Author
-
Tao Liu, Antony Braithwaite, Glenn M. Marshall, Giovanni Perini, Stefan Hüttelmaier, Karen L. MacKenzie, Masoud Vedadi, Cheryl H. Arrowsmith, Peter J. Brown, Johannes H. Schulte, Ygal Haupt, Jason M. Shohet, Quan Zhao, Toby Trahair, Matthew Wong, Bernard Atmadibrata, Bing Liu, Pei Y. Liu, Andrew E. Tee, Rima Al-Awar, Jason W.H. Wong, Giorgio Milazzo, Rebecca C. Poulos, Samuele Gherardi, Daniel Carter, Jessica L. Bell, and Yuting Sun
- Abstract
List of N-Myc-binding gene promoters at which H3K4me3 was not reduced by WDR5 siRNA.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Methods-Figures-Tables from WDR5 Supports an N-Myc Transcriptional Complex That Drives a Protumorigenic Gene Expression Signature in Neuroblastoma
- Author
-
Tao Liu, Antony Braithwaite, Glenn M. Marshall, Giovanni Perini, Stefan Hüttelmaier, Karen L. MacKenzie, Masoud Vedadi, Cheryl H. Arrowsmith, Peter J. Brown, Johannes H. Schulte, Ygal Haupt, Jason M. Shohet, Quan Zhao, Toby Trahair, Matthew Wong, Bernard Atmadibrata, Bing Liu, Pei Y. Liu, Andrew E. Tee, Rima Al-Awar, Jason W.H. Wong, Giorgio Milazzo, Rebecca C. Poulos, Samuele Gherardi, Daniel Carter, Jessica L. Bell, and Yuting Sun
- Abstract
Supplementary Methods-Figures-Tables
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Tables 1-4 from p53 Is a Direct Transcriptional Target of MYCN in Neuroblastoma
- Author
-
Deborah A. Tweddle, John Lunec, Giovanni Perini, Katrina M. Wood, Laura D. Gamble, Samuele Gherardi, Nunzio Iraci, and Lindi Chen
- Abstract
Supplementary Tables 1-4 from p53 Is a Direct Transcriptional Target of MYCN in Neuroblastoma
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Dataset 1 from WDR5 Supports an N-Myc Transcriptional Complex That Drives a Protumorigenic Gene Expression Signature in Neuroblastoma
- Author
-
Tao Liu, Antony Braithwaite, Glenn M. Marshall, Giovanni Perini, Stefan Hüttelmaier, Karen L. MacKenzie, Masoud Vedadi, Cheryl H. Arrowsmith, Peter J. Brown, Johannes H. Schulte, Ygal Haupt, Jason M. Shohet, Quan Zhao, Toby Trahair, Matthew Wong, Bernard Atmadibrata, Bing Liu, Pei Y. Liu, Andrew E. Tee, Rima Al-Awar, Jason W.H. Wong, Giorgio Milazzo, Rebecca C. Poulos, Samuele Gherardi, Daniel Carter, Jessica L. Bell, and Yuting Sun
- Abstract
Genes differentially down- or up-regulated by WDR5 siRNAs by more than 1.5 fold, as shown by Affymetrix microarray, in BE(2)-C neuroblastoma cells 40 hours after siRNA transfection.
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Dataset 5 from WDR5 Supports an N-Myc Transcriptional Complex That Drives a Protumorigenic Gene Expression Signature in Neuroblastoma
- Author
-
Tao Liu, Antony Braithwaite, Glenn M. Marshall, Giovanni Perini, Stefan Hüttelmaier, Karen L. MacKenzie, Masoud Vedadi, Cheryl H. Arrowsmith, Peter J. Brown, Johannes H. Schulte, Ygal Haupt, Jason M. Shohet, Quan Zhao, Toby Trahair, Matthew Wong, Bernard Atmadibrata, Bing Liu, Pei Y. Liu, Andrew E. Tee, Rima Al-Awar, Jason W.H. Wong, Giorgio Milazzo, Rebecca C. Poulos, Samuele Gherardi, Daniel Carter, Jessica L. Bell, and Yuting Sun
- Abstract
List of N-Myc non-binding gene promoters at which H3K4me3 was not reduced by WDR5 siRNA.
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Tables 1-6, Figures 1-4 from N-Myc Regulates Expression of the Detoxifying Enzyme Glutathione Transferase GSTP1, a Marker of Poor Outcome in Neuroblastoma
- Author
-
Michelle Haber, Giovanni Perini, Murray D. Norris, Glenn M. Marshall, Wendy B. London, Lesley J. Ashton, André Oberthuer, Janice Smith, Emanuele Valli, Amanda Russell, Catherine A. Burkhart, Jayne Murray, Samuele Gherardi, and Jamie I. Fletcher
- Abstract
PDF file - 318K
- Published
- 2023
- Full Text
- View/download PDF
14. Reformar la Constitución: ¿la 'cirugía de precisión' obvia la adaptación del resto del ordenamiento? Un análisis a partir del caso italiano
- Author
-
Samuele Gherardi
- Subjects
General Medicine - Abstract
«Una, ninguna y cien mil», son las maneras de reformar el ordenamiento, reza una expresión de Pirandello. Todas ellas pueden llevar a alcanzar el objetivo planteado por el legislador, pero la legitimidad del procedimiento y la puntualidad del ámbito de revisión no son siempre indicativos de un producto final completo, esto es, perfectamente aplicable al sistema jurídico vigente. Para demostrar lo aducido, se trae como ejemplo la revisión constitucional de reducción del número de parlamentarios en Italia de 2020. El intento no es el de estimar si la disposición en cuestión encarna alguna discriminación normativa, sino ilustrar —también de forma visual— la dureza del impacto que puede conllevar una reforma constitucional en su proyección real, particularmente en el ámbito parlamentario.
- Published
- 2021
- Full Text
- View/download PDF
15. La incidencia de los mecanismos parlamentarios en las reformas constitucionales en Italia
- Author
-
Samuele Gherardi
- Published
- 2020
- Full Text
- View/download PDF
16. Reduced menin expression leads to decreased ERα expression and is correlated with the occurrence of human luminal B-like and ER-negative breast cancer subtypes
- Author
-
Romain Teinturier, Chang Xian Zhang, Thomas Bachelot, Laura Corbo, Loay Kassem, Lucie Malbeteau, Yakun Luo, Muriel Le Romancer, Razan Abou Ziki, Isabelle Treilleux, Philippe Bertolino, Samuele Gherardi, Centre de Recherche en Cancérologie de Lyon (UNICANCER/CRCL), Centre Léon Bérard [Lyon]-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre de Lutte contre le Cancer Léon Bérard (CLB), and Manship, Brigitte
- Subjects
Hepatocyte Nuclear Factor 3-alpha ,Cancer Research ,congenital, hereditary, and neonatal diseases and abnormalities ,endocrine system ,endocrine system diseases ,[SDV]Life Sciences [q-bio] ,Breast Neoplasms ,Biology ,Mice ,Breast cancer ,Preclinical Study ,Proto-Oncogene Proteins ,GATA3 ,medicine ,Gene silencing ,Animals ,Humans ,MEN1 ,Promoter Regions, Genetic ,ERα ,Reporter gene ,Gene knockdown ,ESR1 ,Estrogen Receptor alpha ,Menin ,medicine.disease ,[SDV] Life Sciences [q-bio] ,Gene Expression Regulation, Neoplastic ,Oncology ,Cancer research ,MCF-7 Cells ,Female ,FOXA1 ,Luminal subtypes ,Estrogen receptor alpha - Abstract
Purpose Menin, encoded by the MEN1 gene, was recently reported to be involved in breast cancers, though the underlying mechanisms remain elusive. In the current study, we sought to further determine its role in mammary cells. Methods Menin expression in mammary lesions from mammary-specific Men1 mutant mice was detected using immunofluorescence staining. RT-qPCR and western blot were performed to determine the role of menin in ERα expression in human breast cancer cell lines. ChIP-qPCR and reporter gene assays were carried out to dissect the action of menin on the proximal ESR1 promoter. Menin expression in female patients with breast cancer was analyzed and its correlation with breast cancer subtypes was investigated. Results Immunofluorescence staining revealed that early mammary neoplasia in Men1 mutant mice displayed weak ERα expression. Furthermore, MEN1 silencing led to both reduced ESR1 mRNA and ERα protein expression in MCF7 and T47D cells. To further dissect the regulation of ESR1 transcription by menin, we examined whether and in which way menin could regulate the proximal ESR1 promoter, which has not been fully explored. Using ChIP analysis and reporter gene assays covering − 2500 bp to + 2000 bp of the TSS position, we showed that the activity of the proximal ESR1 promoter was markedly reduced upon menin downregulation independently of H3K4me3 status. Importantly, by analyzing the expression of menin in 354 human breast cancers, we found that a lower expression was associated with ER-negative breast cancer (P = 0.041). Moreover, among the 294 ER-positive breast cancer samples, reduced menin expression was not only associated with larger tumors (P = 0.01) and higher SBR grades (P = 0.005) but also with the luminal B-like breast cancer subtype (P = 0.006). Consistent with our clinical data, we demonstrated that GATA3 and FOXA1, co-factors in ESR1 regulation, interact physically with menin in MCF7 cells, and MEN1 knockdown led to altered protein expression of GATA3, the latter being a known marker of the luminal A subtype, in MCF7 cells. Conclusion Taken together, our data provide clues to the important role of menin in ERα regulation and the formation of breast cancer subtypes.
- Published
- 2021
- Full Text
- View/download PDF
17. Men1 disruption in Nkx3.1-deficient mice results in ARlow/CD44+ microinvasive carcinoma development with the dysregulated AR pathway
- Author
-
Razan Abou Ziki, Romain Teinturier, Virginie Firlej, Nicolas Gadot, Muriel Le Romancer, Samuele Gherardi, Myriam Decaussin-Petrucci, Virginie Vlaeminck-Guillem, Remy Bonnavion, Philippe Bertolino, F. Vacherot, Yakun Luo, Isabelle Goddard, Chang Xian Zhang, Centre de Recherche en Cancérologie de Lyon (UNICANCER/CRCL), Centre Léon Bérard [Lyon]-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Résistances Thérapeutiques du Cancer de la Prostate (TRePCa), and Université Paris-Est Créteil Val-de-Marne - Paris 12 (UPEC UP12)
- Subjects
0301 basic medicine ,endocrine system ,Cancer Research ,endocrine system diseases ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,urologic and male genital diseases ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,DU145 ,LNCaP ,Genetics ,medicine ,Gene silencing ,MEN1 ,Molecular Biology ,ComputingMilieux_MISCELLANEOUS ,Gene knockdown ,biology ,CD44 ,medicine.disease ,Androgen receptor ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein - Abstract
Dysregulated androgen receptor (AR) plays a crucial role in prostate cancer (PCa) development, though further factors involved in its regulation remain to be identified. Recently, paradoxical results were reported on the implication of the MEN1 gene in PCa. To dissect its role in prostate luminal cells, we generated a mouse model with inducible Men1 disruption in Nkx3.1-deficient mice in which mouse prostatic intraepithelial neoplasia (mPIN) occur. Prostate glands from mutant and control mice were analyzed pathologically and molecularly; cellular and molecular analyses were carried out in PCa cell lines after MEN1 knockdown (KD) by siRNA. Double-mutant mice developed accelerated mPIN and later displayed microinvasive adenocarcinoma. Markedly, early-stage lesions exhibited a decreased expression of AR and its target genes, accompanied by reduced CK18 and E-cadherin expression, suggesting a shift from a luminal to a dedifferentiated epithelial phenotype. Intriguingly, over 60% of menin-deficient cells expressed CD44 at a later stage. Furthermore, MEN1 KD led to the increase in CD44 expression in PC3 cells re-expressing AR. Menin bound to the proximal AR promoter and regulated AR transcription via the H3K4me3 histone mark. Interestingly, the cell proliferation of AR-dependent cells (LNCaP, 22Rv1, and VCaP), but not of AR-independent cells (DU145, PC3), responded strongly to MEN1 silencing. Finally, menin expression was found reduced in some human PCa. These findings highlight the regulation of the AR promoter by menin and the crosstalk between menin and the AR pathway. Our data could be useful for better understanding the increasingly reported AR-negative/NE-negative subtype of PCa and the mechanisms underlying its development.
- Published
- 2021
- Full Text
- View/download PDF
18. Men1 disruption in Nkx3.1-deficient mice results in AR
- Author
-
Romain, Teinturier, Yakun, Luo, Myriam, Decaussin-Petrucci, Virginie, Vlaeminck-Guillem, Francis, Vacherot, Virginie, Firlej, Rémy, Bonnavion, Razan, Abou Ziki, Samuele, Gherardi, Isabelle, Goddard, Nicolas, Gadot, Philippe, Bertolino, Muriel, Le Romancer, and Chang Xian, Zhang
- Subjects
Homeodomain Proteins ,Male ,Prostatic Intraepithelial Neoplasia ,Prostate ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,Mice ,Hyaluronan Receptors ,Receptors, Androgen ,Proto-Oncogene Proteins ,Animals ,Humans ,Neoplasm Invasiveness ,Cell Proliferation ,Signal Transduction ,Transcription Factors - Abstract
Dysregulated androgen receptor (AR) plays a crucial role in prostate cancer (PCa) development, though further factors involved in its regulation remain to be identified. Recently, paradoxical results were reported on the implication of the MEN1 gene in PCa. To dissect its role in prostate luminal cells, we generated a mouse model with inducible Men1 disruption in Nkx3.1-deficient mice in which mouse prostatic intraepithelial neoplasia (mPIN) occur. Prostate glands from mutant and control mice were analyzed pathologically and molecularly; cellular and molecular analyses were carried out in PCa cell lines after MEN1 knockdown (KD) by siRNA. Double-mutant mice developed accelerated mPIN and later displayed microinvasive adenocarcinoma. Markedly, early-stage lesions exhibited a decreased expression of AR and its target genes, accompanied by reduced CK18 and E-cadherin expression, suggesting a shift from a luminal to a dedifferentiated epithelial phenotype. Intriguingly, over 60% of menin-deficient cells expressed CD44 at a later stage. Furthermore, MEN1 KD led to the increase in CD44 expression in PC3 cells re-expressing AR. Menin bound to the proximal AR promoter and regulated AR transcription via the H3K4me3 histone mark. Interestingly, the cell proliferation of AR-dependent cells (LNCaP, 22Rv1, and VCaP), but not of AR-independent cells (DU145, PC3), responded strongly to MEN1 silencing. Finally, menin expression was found reduced in some human PCa. These findings highlight the regulation of the AR promoter by menin and the crosstalk between menin and the AR pathway. Our data could be useful for better understanding the increasingly reported AR-negative/NE-negative subtype of PCa and the mechanisms underlying its development.
- Published
- 2020
19. Due modelli costituzionali per governare l’emergenza. Italia e Spagna alla prova del Coronavirus
- Author
-
Baldoni, Diego and Samuele, Gherardi
- Subjects
Coronavirus ,Italy ,Spain ,Public comparative law, System of sources, Italy, Spain, State of emergency, Coronavirus ,System of sources ,Public comparative law ,State of emergency - Published
- 2020
20. Menin regulates Inhbb expression through an Akt/Ezh2-mediated H3K27 histone modification
- Author
-
Martine Cordier-Bussat, Philippe Bertolino, Ana Hennino, Delphine Goehrig, Chang X. Zhang, Ivan Mikaelian, Romain Teinturier, Doriane Ripoche, Samuele Gherardi, and Marie Chanal
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,endocrine system ,endocrine system diseases ,Biophysics ,macromolecular substances ,Methylation ,Biochemistry ,Histones ,03 medical and health sciences ,0302 clinical medicine ,Structural Biology ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Gene expression ,Genetics ,Animals ,Enhancer of Zeste Homolog 2 Protein ,MEN1 ,Epigenetics ,Phosphorylation ,Promoter Regions, Genetic ,Molecular Biology ,Inhibin-beta Subunits ,Mice, Knockout ,Polycomb Repressive Complex 1 ,Regulation of gene expression ,biology ,Lysine ,EZH2 ,Polycomb Repressive Complex 2 ,Fibroblasts ,Embryo, Mammalian ,Mice, Inbred C57BL ,INHBB ,030104 developmental biology ,Histone ,Gene Expression Regulation ,Genetic Loci ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,H3K4me3 ,Proto-Oncogene Proteins c-akt ,Protein Binding ,Signal Transduction - Abstract
Although Men1 is a well-known tumour suppressor gene, little is known about the functions of Menin, the protein it encodes for. Since few years, numerous publications support a major role of Menin in the control of epigenetics gene regulation. While Menin interaction with MLL complex favours transcriptional activation of target genes through H3K4me3 marks, Menin also represses gene expression via mechanisms involving the Polycomb repressing complex (PRC). Interestingly, Ezh2, the PRC-methyltransferase that catalyses H3K27me3 repressive marks and Menin have been shown to co-occupy a large number of promoters. However, lack of binding between Menin and Ezh2 suggests that another member of the PRC complex is mediating this indirect interaction. Having found that ActivinB - a TGFβ superfamily member encoded by the Inhbb gene - is upregulated in insulinoma tumours caused by Men1 invalidation, we hypothesize that Menin could directly participate in the epigenetic-repression of Inhbb gene expression. Using Animal model and cell lines, we report that loss of Menin is directly associated with ActivinB-induced expression both in vivo and in vitro. Our work further reveals that ActivinB expression is mediated through a direct modulation of H3K27me3 marks on the Inhbb locus in Menin-KO cell lines. More importantly, we show that Menin binds on the promoter of Inhbb gene where it favours the recruitment of Ezh2 via an indirect mechanism involving Akt-phosphorylation. Our data suggests therefore that Menin could take an important part to the Ezh2-epigenetic repressive landscape in many cells and tissues through its capacity to modulate Akt phosphorylation.
- Published
- 2017
- Full Text
- View/download PDF
21. Foxa2, a novel protein partner of the tumour suppressor menin, is deregulated in mouse and humanMEN1glucagonomas
- Author
-
Marie-Christine Vantyghem, Rémy Bonnavion, Emmanuelle Leteurtre, Rui Du, François Pattou, Romain Teinturier, Chang Xian Zhang, Samuele Gherardi, Run Yu, Jieli Lu, Philippe Bertolino, and Martine Cordier-Bussat
- Subjects
0301 basic medicine ,endocrine system ,endocrine system diseases ,Regulator ,Enteroendocrine cell ,respiratory system ,Biology ,medicine.disease ,Pathology and Forensic Medicine ,law.invention ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,law ,030220 oncology & carcinogenesis ,embryonic structures ,medicine ,Cancer research ,Suppressor ,MEN1 ,FOXA2 ,Multiple endocrine neoplasia ,Gene ,reproductive and urinary physiology ,Immunostaining - Abstract
Foxa2, known as one of the pioneer factors, plays a crucial role in islet development and endocrine functions. Its expression and biological functions are regulated by various factors, including, in particular, insulin and glucagon. However, its expression and biological role in adult pancreatic α-cells remain elusive. In the current study, we showed that Foxa2 was overexpressed in islets from α-cell-specific Men1 mutant mice, at both the transcriptional level and the protein level. More importantly, immunostaining analyses showed its prominent nuclear accumulation, specifically in α-cells, at a very early stage after Men1 disruption. Similar nuclear FOXA2 expression was also detected in a substantial proportion (12/19) of human multiple endocrine neoplasia type 1 (MEN1) glucagonomas. Interestingly, our data revealed an interaction between Foxa2 and menin encoded by the Men1 gene. Furthermore, using several approaches, we demonstrated the relevance of this interaction in the regulation of two tested Foxa2 target genes, including the autoregulation of the Foxa2 promoter by Foxa2 itself. The current study establishes menin, a novel protein partner of Foxa2, as a regulator of Foxa2, the biological functions of which extend beyond the pancreatic endocrine cells. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
- Published
- 2017
- Full Text
- View/download PDF
22. Corrigendum to: 'Transcriptional and epigenetic analyses of the DMD locus reveal novel cis-acting DNA elements that govern muscle dystrophin expression'. [Biochim. Biophys. Acta Gene Regul. Mech. 2017 Nov;1860(11):1138–1147.]
- Author
-
C. Scotton, H. Osman, Alessandra Recchia, Lucia Morandi, Alessandra Ferlini, Marcella Neri, Pia Bernasconi, Paolo Pigini, Lorenzo Maggi, Chiara Passarelli, Matteo Bovolenta, Marina Mora, Annarita Armaroli, Maria Sofia Falzarano, Samuele Gherardi, Rita Selvatici, Giovanni Perini, and Francesca Gualandi
- Subjects
Genetics ,Biophysics ,Locus (genetics) ,Biology ,Biochemistry ,Cis acting ,chemistry.chemical_compound ,chemistry ,Structural Biology ,biology.protein ,Epigenetics ,Dystrophin ,Molecular Biology ,Gene ,DNA - Published
- 2020
- Full Text
- View/download PDF
23. ABCC Multidrug Transporters in Childhood Neuroblastoma: Clinical and Biological Effects Independent of Cytotoxic Drug Efflux
- Author
-
Wendy B. London, Amanda J. Russell, Chin Kiat Kwek, Emanuele Valli, Claudia Flemming, Antonio Porro, Jamie I. Fletcher, Lesley J. Ashton, Michelle Haber, Jayne Murray, Janice Smith, Susan L. Cohn, Murray D. Norris, Samuele Gherardi, Giovanni Perini, Alan C. Sartorelli, Michelle J. Henderson, Manfred Schwab, Marcia A. Munoz, Glenn M. Marshall, Chengyuan Xue, Nunzio Iraci, and Allen Buxton
- Subjects
Male ,Cancer Research ,Pathology ,Time Factors ,Kaplan-Meier Estimate ,Polymerase Chain Reaction ,Mice ,Neuroblastoma ,0302 clinical medicine ,Cell Movement ,Recurrence ,Odds Ratio ,ABCC TRANSPORTERS ,NEUROBLASTOMA ,CHEMORESISTANCE ,GENE TRANSCRIPTION ,MRP1-KO MICE ,Prospective Studies ,RNA, Small Interfering ,Child ,Oncogene Proteins ,N-Myc Proto-Oncogene Protein ,0303 health sciences ,biology ,Hazard ratio ,Nuclear Proteins ,Cell Differentiation ,Prognosis ,Drug Resistance, Multiple ,Up-Regulation ,3. Good health ,Gene Expression Regulation, Neoplastic ,Oncology ,Child, Preschool ,030220 oncology & carcinogenesis ,ABCC3 ,ABCC1 ,Female ,Multidrug Resistance-Associated Proteins ,Childhood Neuroblastoma ,medicine.medical_specialty ,Adolescent ,Blotting, Western ,Down-Regulation ,Antineoplastic Agents ,Mice, Transgenic ,ABCC4 ,Transfection ,Disease-Free Survival ,Young Adult ,03 medical and health sciences ,Predictive Value of Tests ,Cell Line, Tumor ,Internal medicine ,medicine ,Animals ,Humans ,Gene Silencing ,Adverse effect ,Proportional Hazards Models ,030304 developmental biology ,Cell growth ,Editorials ,Infant ,medicine.disease ,Disease Models, Animal ,Pyrimidines ,Endocrinology ,Drug Resistance, Neoplasm ,biology.protein ,Pyrazoles - Abstract
Background Although the prognostic value of the ATP-binding cassette, subfamily C (ABCC) transporters in childhood neuroblastoma is usually attributed to their role in cytotoxic drug efflux, certain observations have suggested that these multidrug transporters might contribute to the malignant phenotype independent of cytotoxic drug efflux. Methods A v-myc myelocytomatosis viral related oncogene, neuroblastoma derived (MYCN)-driven transgenic mouse neuroblastoma model was crossed with an Abcc1-deficient mouse strain (658 hMYCN1/−, 205 hMYCN+/1 mice) or, alternatively, treated with the ABCC1 inhibitor, Reversan (n = 20). ABCC genes were suppressed using short interfering RNA or overexpressed by stable transfection in neuroblastoma cell lines BE(2)-C, SH-EP, and SH-SY5Y, which were then assessed for wound closure ability, clonogenic capacity, morphological differentiation, and cell growth. Real-time quantitative polymerase chain reaction was used to examine the clinical significance of ABCC family gene expression in a large prospectively accrued cohort of patients (n = 209) with primary neuroblastomas. Kaplan-Meier survival analysis and Cox regression were used to test for associations with event-free and overall survival. Except where noted, all statistical tests were two-sided. Results Inhibition of ABCC1 statistically significantly inhibited neuroblastoma development in hMYCN transgenic mice (mean age for palpable tumor: treated mice, 47.2 days; control mice, 41.9 days; hazard ratio [HR] = 9.3, 95% confidence interval [CI] = 2.65 to 32; P < .001). Suppression of ABCC1 in vitro inhibited wound closure (P < .001) and clonogenicity (P = .006); suppression of ABCC4 enhanced morphological differentiation (P < .001) and inhibited cell growth (P < .001). Analysis of 209 neuroblastoma patient tumors revealed that, in contrast with ABCC1 and ABCC4, low rather than high ABCC3 expression was associated with reduced event-free survival (HR of recurrence or death = 2.4, 95% CI = 1.4 to 4.2; P = .001), with 23 of 53 patients with low ABCC3 expression experiencing recurrence or death compared with 31 of 155 patients with high ABCC3. Moreover, overexpression of ABCC3 in vitro inhibited neuroblastoma cell migration (P < .001) and clonogenicity (P = .03). The combined expression of ABCC1, ABCC3, and ABCC4 was associated with patients having an adverse event, such that of the 12 patients with the "poor prognosis” expression pattern, 10 experienced recurrence or death (HR of recurrence or death = 12.3, 95% CI = 6 to 27; P < .001). Conclusion ABCC transporters can affect neuroblastoma biology independently of their role in chemotherapeutic drug efflux, enhancing their potential as targets for therapeutic intervention
- Published
- 2017
24. Transcriptional and epigenetic analyses of the DMD locus reveal novel cisâacting DNA elements that govern muscle dystrophin expression
- Author
-
Paolo Pigini, Maria Sofia Falzarano, Alessandra Ferlini, Marina Mora, C. Scotton, Pia Bernasconi, H. Osman, Lucia Morandi, Matteo Bovolenta, Lorenzo Maggi, Francesca Gualandi, Giovanni Perini, Samuele Gherardi, Alessandra Recchia, Marcella Neri, Rita Selvatici, Chiara Passarelli, Annarita Armaroli, Gherardi, Samuele, Bovolenta, Matteo, Passarelli, Chiara, Falzarano, Maria Sofia, Pigini, Paolo, Scotton, Chiara, Neri, Marcella, Armaroli, Annarita, Osman, Hana, Selvatici, Rita, Gualandi, Francesca, Recchia, Alessandra, Mora, Marina, Bernasconi, Pia, Maggi, Lorenzo, Morandi, Lucia, Ferlini, Alessandra, and Perini, Giovanni
- Subjects
Adult ,0301 basic medicine ,musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,Adolescent ,Biophysics ,Locus (genetics) ,Regulatory Sequences, Nucleic Acid ,Biology ,Gene mutation ,Biochemistry ,Epigenesis, Genetic ,NO ,Dystrophin ,Mice ,Young Adult ,03 medical and health sciences ,Duchenne Muscular Dystrophy (DMD) ,Transcriptional regulation ,Structural Biology ,Utrophin ,Becker Muscular Dystrophy (BMD) ,Genetics ,Animals ,Humans ,Chromosome Conformation Capture (3C) ,RNA pol II pausing ,Molecular Biology ,Child ,Muscle, Skeletal ,Gene ,Cells, Cultured ,Regulation of gene expression ,Chromatin ,Muscular Dystrophy, Duchenne ,030104 developmental biology ,Gene Expression Regulation ,Biophysic ,Child, Preschool ,Mutation ,biology.protein ,HeLa Cells - Abstract
The dystrophin gene (DMD) is the largest gene in the human genome, mapping on the Xp21 chromosome locus. It spans 2.2 Mb and accounts for approximately 0,1% of the entire human genome. Mutations in this gene cause Duchenne and Becker Muscular Dystrophy, X-linked Dilated Cardiomyopathy, and other milder muscle phenotypes. Beside the remarkable number of reports describing dystrophin gene expression and the pathogenic consequences of the gene mutations in dystrophinopathies, the full scenario of the DMD transcription dynamics remains however, poorly understood. Considering that the full transcription of the DMD gene requires about 16 h, we have investigated the activity of RNA Polymerase II along the entire DMD locus within the context of specific chromatin modifications using a variety of chromatin-based techniques. Our results unveil a surprisingly powerful processivity of the RNA polymerase II along the entire 2.2 Mb of the DMD locus with just one site of pausing around intron 52. We also discovered epigenetic marks highlighting the existence of four novel cisâDNA elements, two of which, located within intron 34 and exon 45, appear to govern the architecture of the DMD chromatin with implications on the expression levels of the muscle dystrophin mRNA. Overall, our findings provide a global view on how the entire DMD locus is dynamically transcribed by the RNA pol II and shed light on the mechanisms involved in dystrophin gene expression control, which can positively impact on the optimization of the novel ongoing therapeutic strategies for dystrophinopathies.
- Published
- 2017
25. Physical Interaction between MYCN Oncogene and Polycomb Repressive Complex 2 (PRC2) in Neuroblastoma
- Author
-
Daisy Corvetta, Giovanni Perini, Arturo Sala, Izabela Piotrowska, Emanuele Valli, Cosimo Walter D'Acunto, Samuele Gherardi, Olesya Chayka, and Sandra Cantilena
- Subjects
0303 health sciences ,biology ,Tumor suppressor gene ,Clusterin ,EZH2 ,Cell Biology ,medicine.disease_cause ,Biochemistry ,Molecular biology ,Cell biology ,Chromatin ,03 medical and health sciences ,0302 clinical medicine ,RNA interference ,030220 oncology & carcinogenesis ,biology.protein ,medicine ,Epigenetics ,PRC2 ,Carcinogenesis ,neoplasms ,Molecular Biology ,030304 developmental biology - Abstract
CLU (clusterin) is a tumor suppressor gene that we have previously shown to be negatively modulated by the MYCN proto-oncogene, but the mechanism of repression was unclear. Here, we show that MYCN inhibits the expression of CLU by direct interaction with the non-canonical E box sequence CACGCG in the 5′-flanking region. Binding of MYCN to the CLU gene induces bivalent epigenetic marks and recruitment of repressive proteins such as histone deacetylases and Polycomb members. MYCN physically binds in vitro and in vivo to EZH2, a component of the Polycomb repressive complex 2, required to repress CLU. Notably, EZH2 interacts with the Myc box domain 3, a segment of MYC known to be essential for its transforming effects. The expression of CLU can be restored in MYCN-amplified cells by epigenetic drugs with therapeutic results. Importantly, the anticancer effects of the drugs are ablated if CLU expression is blunted by RNA interference. Our study implies that MYC tumorigenesis can be effectively antagonized by epigenetic drugs that interfere with the recruitment of chromatin modifiers at repressive E boxes of tumor suppressor genes such as CLU.
- Published
- 2013
- Full Text
- View/download PDF
26. Epigenetic role of the N-MYC/LSD1 complex in Neuroblastoma
- Author
-
SORRENTINO, MARIA CRISTINA, AMENTE, STEFANO, AMBROSIO, SUSANNA, LANIA, LUIGI, MAJELLO, BARBARA, Giorgio Milazzo, Cinzia Raimondo, Samuele Gherardi, Giovanni Perini, FISV, Sorrentino, MARIA CRISTINA, Giorgio, Milazzo, Amente, Stefano, Cinzia, Raimondo, Ambrosio, Susanna, Samuele, Gherardi, Lania, Luigi, Giovanni, Perini, and Majello, Barbara
- Published
- 2014
27. Epigenetic role of the MYCN/LSD1 complex iin Neuroblastoma
- Author
-
AMENTE, STEFANO, SORRENTINO, MARIA CRISTINA, AMBROSIO, SUSANNA, LANIA, LUIGI, MAJELLO, BARBARA, Giorgio Milazzo, Cinzia Raimondo, Samuele Gherardi, Giovanni Perini, ANR, Amente, Stefano, Giorgio, Milazzo, Sorrentino, MARIA CRISTINA, Cinzia, Raimondo, Ambrosio, Susanna, Samuele, Gherardi, Lania, Luigi, Giovanni, Perini, and Majello, Barbara
- Published
- 2014
28. Foxa2, a novel protein partner of the tumour suppressor menin, is deregulated in mouse and human MEN1 glucagonomas
- Author
-
Rémy, Bonnavion, Romain, Teinturier, Samuele, Gherardi, Emmanuelle, Leteurtre, Run, Yu, Martine, Cordier-Bussat, Rui, Du, François, Pattou, Marie-Christine, Vantyghem, Philippe, Bertolino, Jieli, Lu, and Chang Xian, Zhang
- Subjects
Gene Expression Regulation, Neoplastic ,Pancreatic Neoplasms ,Proto-Oncogene Proteins ,Hepatocyte Nuclear Factor 3-beta ,Multiple Endocrine Neoplasia Type 1 ,Tumor Cells, Cultured ,Animals ,Glucagonoma ,Humans ,Mice, Transgenic ,Promoter Regions, Genetic ,Transfection ,Neoplasm Proteins - Abstract
Foxa2, known as one of the pioneer factors, plays a crucial role in islet development and endocrine functions. Its expression and biological functions are regulated by various factors, including, in particular, insulin and glucagon. However, its expression and biological role in adult pancreatic α-cells remain elusive. In the current study, we showed that Foxa2 was overexpressed in islets from α-cell-specific Men1 mutant mice, at both the transcriptional level and the protein level. More importantly, immunostaining analyses showed its prominent nuclear accumulation, specifically in α-cells, at a very early stage after Men1 disruption. Similar nuclear FOXA2 expression was also detected in a substantial proportion (12/19) of human multiple endocrine neoplasia type 1 (MEN1) glucagonomas. Interestingly, our data revealed an interaction between Foxa2 and menin encoded by the Men1 gene. Furthermore, using several approaches, we demonstrated the relevance of this interaction in the regulation of two tested Foxa2 target genes, including the autoregulation of the Foxa2 promoter by Foxa2 itself. The current study establishes menin, a novel protein partner of Foxa2, as a regulator of Foxa2, the biological functions of which extend beyond the pancreatic endocrine cells. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John WileySons, Ltd.
- Published
- 2015
29. WDR5 Supports an N-Myc Transcriptional Complex That Drives a Protumorigenic Gene Expression Signature in Neuroblastoma
- Author
-
Antony W. Braithwaite, Rima Al-awar, Johannes H. Schulte, Giorgio Milazzo, Daniel R. Carter, Jason M. Shohet, Ygal Haupt, Andrew E. Tee, Bing Bing Liu, Stefan Hüttelmaier, Quan Zhao, Giovanni Perini, Masoud Vedadi, Toby Trahair, Glenn M. Marshall, Bernard Atmadibrata, Pei Y. Liu, Samuele Gherardi, Jessica L. Bell, Yuting Sun, Matthew Wong, Jason W. H. Wong, Cheryl H. Arrowsmith, Karen L. MacKenzie, Rebecca C. Poulos, Peter Brown, Tao Liu, Sun, Yuting, Bell, Jessica L, Carter, Daniel, Gherardi, Samuele, Poulos, Rebecca C, Milazzo, Giorgio, Wong, Jason W H, Al-Awar, Rima, Tee, Andrew E, Liu, Pei Y, Liu, Bing, Atmadibrata, Bernard, Wong, Matthew, Trahair, Toby, Zhao, Quan, Shohet, Jason M, Haupt, Ygal, Schulte, Johannes H, Brown, Peter J, Arrowsmith, Cheryl H, Vedadi, Masoud, Mackenzie, Karen L, Hüttelmaier, Stefan, Perini, Giovanni, Marshall, Glenn M, Braithwaite, Antony, and Liu, Tao
- Subjects
Cancer Research ,Transcription, Genetic ,Carcinogenesis ,Genes, myc ,Mice, Transgenic ,Cell Growth Processes ,Methylation ,Histones ,Proto-Oncogene Proteins c-myc ,Mice ,Neuroblastoma ,Gene expression ,medicine ,WDR5 ,Animals ,Humans ,Promoter Regions, Genetic ,neoplasms ,biology ,HEK 293 cells ,Intracellular Signaling Peptides and Proteins ,Promoter ,Proto-Oncogene Proteins c-mdm2 ,Histone-Lysine N-Methyltransferase ,medicine.disease ,Rats ,Up-Regulation ,Histone ,HEK293 Cells ,Oncology ,biology.protein ,Cancer research ,Mdm2 ,Tumor Suppressor Protein p53 ,Transcriptome ,N-Myc , WDR5, Neuroblastoma, Transcriptional regulation, histone acetylase epigenetics, chromatin ,N-Myc - Abstract
MYCN gene amplification in neuroblastoma drives a gene expression program that correlates strongly with aggressive disease. Mechanistically, trimethylation of histone H3 lysine 4 (H3K4) at target gene promoters is a strict prerequisite for this transcriptional program to be enacted. WDR5 is a histone H3K4 presenter that has been found to have an essential role in H3K4 trimethylation. For this reason, in this study, we investigated the relationship between WDR5-mediated H3K4 trimethylation and N-Myc transcriptional programs in neuroblastoma cells. N-Myc upregulated WDR5 expression in neuroblastoma cells. Gene expression analysis revealed that WDR5 target genes included those with MYC-binding elements at promoters such as MDM2. We showed that WDR5 could form a protein complex at the MDM2 promoter with N-Myc, but not p53, leading to histone H3K4 trimethylation and activation of MDM2 transcription. RNAi-mediated attenuation of WDR5 upregulated expression of wild-type but not mutant p53, an effect associated with growth inhibition and apoptosis. Similarly, a small-molecule antagonist of WDR5 reduced N-Myc/WDR5 complex formation, N-Myc target gene expression, and cell growth in neuroblastoma cells. In MYCN-transgenic mice, WDR5 was overexpressed in precancerous ganglion and neuroblastoma cells compared with normal ganglion cells. Clinically, elevated levels of WDR5 in neuroblastoma specimens were an independent predictor of poor overall survival. Overall, our results identify WDR5 as a key cofactor for N-Myc–regulated transcriptional activation and tumorigenesis and as a novel therapeutic target for MYCN-amplified neuroblastomas. Cancer Res; 75(23); 5143–54. ©2015 AACR.
- Published
- 2015
30. Omics approach and novel biostatistic tools identified RPL3L as potential genetic modifier of clinical severity in female carriers of Duchenne muscle dystrophy
- Author
-
C. Scotton, Eugenio Mercuri, Chiara Passarelli, Anton Yuryev, Chiara Scapoli, Annarita Armaroli, Graziano Pesole, L. Wenyan, Enrico Bertini, Francesca Gualandi, Samuele Gherardi, Alessandra Ferlini, Marika Pane, Adele D'Amico, Elena Schwartz, Alberto Carrieri, Matteo Bovolenta, F. Mingyan, F. Di Raimo, and Marcella Neri
- Subjects
Genetics ,Neurology ,Pediatrics, Perinatology and Child Health ,Clinical severity ,Neurology (clinical) ,Muscle dystrophy ,Computational biology ,Biology ,Omics ,Genetics (clinical) ,NO - Published
- 2015
31. SKP2 is a direct transcriptional target of MYCN and a potential therapeutic target in neuroblastoma
- Author
-
Elaine Willmore, Giovanni Perini, Deborah A. Tweddle, Laura Evans, Samuele Gherardi, Giorgio Milazzo, Lindi Chen, David R. Newell, Evans, Laura, Chen, Lindi, Milazzo, Giorgio, Gherardi, Samuele, Perini, Giovanni, Willmore, Elaine, Newell, David R., and Tweddle, Deborah A.
- Subjects
Cancer Research ,Time Factors ,Time Factor ,Transcription, Genetic ,G1 arrest ,Apoptosis ,Biology ,Transfection ,E-Box Elements ,Neuroblastoma ,Genes, Reporter ,Cell Line, Tumor ,MYCN ,medicine ,SKP2 ,G1 Phase Cell Cycle Checkpoint ,Humans ,E-Box Element ,RNA, Messenger ,Promoter Regions, Genetic ,S-Phase Kinase-Associated Proteins ,neoplasms ,Cell Proliferation ,Nuclear Protein ,Oncogene Proteins ,N-Myc Proto-Oncogene Protein ,Reporter gene ,Gene knockdown ,Binding Sites ,Cell growth ,Medicine (all) ,Binding Site ,Oncogene Protein ,Nuclear Proteins ,Apoptosi ,Cell cycle ,medicine.disease ,G1 Phase Cell Cycle Checkpoints ,Gene Expression Regulation, Neoplastic ,Oncology ,Ubiquitin ligase complex ,Mutation ,Cancer research ,RNA Interference ,Chromatin immunoprecipitation ,S-Phase Kinase-Associated Protein ,Human - Abstract
SKP2 is the substrate recognition subunit of the ubiquitin ligase complex which targets p27(KIP1) for degradation. Induced at the G1/S transit of the cell cycle, SKP2 is frequently overexpressed in human cancers and contributes to malignancy. We previously identified SKP2 as a possible MYCN target gene and hence hypothesise that SKP2 is a potential therapeutic target in MYCN amplified disease. A positive correlation was identified between MYCN activity and SKP2 mRNA expression in Tet21N MYCN-regulatable cells and a panel of MYCN amplified and non-amplified neuroblastoma cell lines. In chromatin immunoprecipitation and reporter gene assays, MYCN bound directly to E-boxes within the SKP2 promoter and induced transcriptional activity which was decreased by the removal of MYCN and E-box mutation. Although SKP2 knockdown inhibited cell growth in both MYCN amplified and non-amplified cells, cell cycle arrest and apoptosis were induced only in non-MYCN amplified neuroblastoma cells. In conclusion these data identify SKP2 as a direct transcriptional target of MYCN and supports SKP2 as a potential therapeutic target in neuroblastoma.
- Published
- 2015
32. Abstract 2450: MYCN and TFAP4 promote neuroblastoma malignancy by cooperating in the regulation a subset of target genes involved in cancer cell growth and metastasis
- Author
-
Belamy B. Cheung, Bing Liu, Amanda J. Russell, Chengyuan Xue, Tao Liu, Murray D. Norris, Giorgio Milazzo, Laura D. Gamble, Glenn M. Marshall, Jessica Koach, Denise M. Yu, Samuele Gherardi, Michelle Haber, and Giovanni Perini
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Cancer ,Biology ,medicine.disease ,Metastasis ,03 medical and health sciences ,030104 developmental biology ,Tumor progression ,Enhancer binding ,Internal medicine ,Neuroblastoma ,Cancer cell ,medicine ,Cancer research ,TFAP4 ,neoplasms ,Transcription factor - Abstract
Amplification of the MYCN oncogene, a member of the MYC family of transcriptional regulators, is one of the most powerful prognostic markers identified for poor outcome in neuroblastoma, the most common extracranial solid cancer in childhood. While MYCN has been established as a key driver of malignancy in neuroblastoma, the underlying molecular mechanisms are poorly understood. Transcription factor activating enhancer binding protein-4 (TFAP4), which plays important roles in cancer progression, has been reported to be a direct transcriptional target of MYC. In this study, we have shown that high expression of TFAP4 in primary neuroblastoma patients is associated with poor clinical outcome and furthermore that siRNA-mediated suppression of TFAP4 in MYCN-expressing neuroblastoma cells impaired migration and colony formation, and led to an increased proportion of cells in G1/S phase of the cell cycle. Chromatin immunoprecipitation and luciferase reporter assays demonstrated that TFAP4 expression is positively regulated by MYCN through direct promoter binding. In addition, when MYCN was overexpressed in neuroblastoma cells, TFAP4 was required for the observed increase in cell migration. Microarray analysis identified genes regulated by both MYCN and TFAP4 in neuroblastoma cells, including Phosphoribosyl-pyrophosphate synthetase-2 (PRPS2) and Syndecan-1 (SDC1), which are involved in cancer cell proliferation and metastasis. Overall this study unveils a complex regulatory circuit in which MYCN by elevating TFAP4 expression, cooperates with it to control a specific set of genes involved in tumor progression. These findings highlight the existence of a MYCN-TFAP4 axis in MYCN-driven neuroblastoma as well as identifying relevant therapeutic targets for aggressive forms of this disease. Citation Format: Chengyuan Xue, Denise M. Yu, Samuele Gherardi, Jessica Koach, Giorgio Milazzo, Laura Gamble, Bing Liu, Amanda Russell, Tao Liu, Belamy B. Cheung, Glenn M. Marshall, Giovanni Perini, Michelle Haber, Murray D. Norris. MYCN and TFAP4 promote neuroblastoma malignancy by cooperating in the regulation a subset of target genes involved in cancer cell growth and metastasis. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2450.
- Published
- 2016
- Full Text
- View/download PDF
33. The DMD Locus Harbours Multiple Long Non-Coding RNAs Which Orchestrate and Control Transcription of Muscle Dystrophin mRNA Isoforms
- Author
-
Alessandra Ferlini, Emanuele Valli, Matteo Bovolenta, Marcella Neri, Paola Rimessi, Maria Sofia Falzarano, Daniela Erriquez, C. Scotton, Francesca Gualandi, Giovanni Perini, Samuele Gherardi, S. Brioschi, M. Fabris, Bovolenta M., Erriquez D., Valli E., Brioschi S., Scotton C., Neri M., Falzarano M.S., Gherardi S., Fabris M., Rimessi P., Gualandi F., Perini G., and Ferlini A.
- Subjects
Gene isoform ,Male ,musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,Transcription, Genetic ,LONG NON CODING RNAS ,Duchenne muscular dystrophy ,DNA transcription ,lcsh:Medicine ,Duchenne Muscular Dystrophy ,Biology ,DUCHENNE MUSCLE DYSTROPHY ,ALTERNATIVE PROMOTERS ,Molecular Genetics ,03 medical and health sciences ,0302 clinical medicine ,RNA interference ,RNA Isoforms ,TRANSCRIPTION REGULATION ,Utrophin ,medicine ,Genetics ,Humans ,DYSTROPHIN ,Gene Regulation ,Muscle, Skeletal ,lcsh:Science ,Gene ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,lcsh:R ,Human Genetics ,X-Linked ,medicine.disease ,Long non-coding RNA ,RNA processing ,biology.protein ,Ectopic expression ,Female ,RNA, Long Noncoding ,lcsh:Q ,Gene expression ,Dystrophin ,030217 neurology & neurosurgery ,Research Article - Abstract
The 2.2 Mb long dystrophin (DMD) gene, the largest gene in the human genome, corresponds to roughly 0.1% of the entire human DNA sequence. Mutations in this gene cause Duchenne muscular dystrophy and other milder X-linked, recessive dystrophinopathies. Using a custom-made tiling array, specifically designed for the DMD locus, we identified a variety of novel long non-coding RNAs (lncRNAs), both sense and antisense oriented, whose expression profiles mirror that of DMD gene. Importantly, these transcripts are intronic in origin and specifically localized to the nucleus and are transcribed contextually with dystrophin isoforms or primed by MyoD-induced myogenic differentiation. Furthermore, their forced ectopic expression in both human muscle and neuronal cells causes a specific and negative regulation of endogenous dystrophin full length isoforms and significantly down-regulate the activity of a luciferase reporter construct carrying the minimal promoter regions of the muscle dystrophin isoform. Consistent with this apparently repressive role, we found that, in muscle samples of dystrophinopathic female carriers, lncRNAs expression levels inversely correlate with those of muscle full length DMD isoforms. Overall these findings unveil an unprecedented complexity of the transcriptional pattern of the DMD locus and reveal that DMD lncRNAs may contribute to the orchestration and homeostasis of the muscle dystrophin expression pattern by either selective targeting and down-modulating the dystrophin promoter transcriptional activity.
- Published
- 2012
34. N-Myc regulates expression of the detoxifying enzyme glutathione transferase GSTP1, a marker of poor outcome in neuroblastoma
- Author
-
Glenn M. Marshall, Wendy B. London, Jamie I. Fletcher, Samuele Gherardi, Catherine Burkhart, Janice Smith, André Oberthuer, Michelle Haber, Jayne Murray, Giovanni Perini, Lesley J. Ashton, Emanuele Valli, Murray D. Norris, Amanda J. Russell, Fletcher J.I., Gherardi S., Murray J., Burkhart C.A., Russell A., Valli E., Smith J., Oberthuer A., Ashton L.J., London W.B., Marshall G.M., Norris M.D., Perini G., and Haber M.
- Subjects
Male ,Cancer Research ,ATP-binding cassette transporter ,Mice, Transgenic ,Biology ,N-Myc Proto-Oncogene Protein ,Cohort Studies ,GSTP1 ,Mice ,Neuroblastoma ,ABC TRANSPORTERS ,Transcriptional regulation ,medicine ,Animals ,Humans ,neoplasms ,Regulation of gene expression ,Oncogene Proteins ,N-MYC ,MYCN AMPLIFICATION ,Nuclear Proteins ,medicine.disease ,Prognosis ,Gene Expression Regulation, Neoplastic ,Oncology ,Glutathione S-Transferase pi ,Cancer research ,Female ,Efflux ,N-Myc - Abstract
Amplification of the transcription factor MYCN is associated with poor outcome and a multidrug-resistant phenotype in neuroblastoma. N-Myc regulates the expression of several ATP-binding cassette (ABC) transporter genes, thus affecting global drug efflux. Because these transporters do not confer resistance to several important cytotoxic agents used to treat neuroblastoma, we explored the prognostic significance and transcriptional regulation of the phase II detoxifying enzyme, glutathione S-transferase P1 (GSTP1). Using quantitative real-time PCR, GSTP1 gene expression was assessed in a retrospective cohort of 51 patients and subsequently in a cohort of 207 prospectively accrued primary neuroblastomas. These data along with GSTP1 expression data from an independent microarray study of 251 neuroblastoma samples were correlated with established prognostic indicators and disease outcome. High levels of GSTP1 were associated with decreased event-free and overall survival in all three cohorts. Multivariable analyses, including age at diagnosis, tumor stage, and MYCN amplification status, were conducted on the two larger cohorts, independently showing the prognostic significance of GSTP1 expression levels in this setting. Mechanistic investigations revealed that GSTP1 is a direct transcriptional target of N-Myc in neuroblastoma cells. Together, our findings reveal that N-Myc regulates GSTP1 along with ABC transporters that act to control drug metabolism and efflux. Furthermore, they imply that strategies to jointly alter these key multidrug resistance mechanisms may have therapeutic implications to manage neuroblastomas and other malignancies driven by amplified Myc family genes. Cancer Res; 72(4); 845–53. ©2011 AACR.
- Published
- 2011
35. c-MYC oncoprotein dictates transcriptional profiles of ATP-binding cassette transporter genes in Chronic Myelogenous Leukemia CD34+ hematopoietic progenitor cells
- Author
-
Daniel Diolaiti, Simona Soverini, Murray D. Norris, Giovanni Perini, Emanuele Valli, Carolina Terragna, Michelle Haber, Giovanni Martinelli, Nunzio Iraci, Thea Kalebic, Antonio Porro, Chiara Perrod, Roberto Bernardoni, Michele Baccarani, Samuele Gherardi, Sandra Durante, A. Porro, N. Iraci, S. Soverini, D. Diolaiti, S. Gherardi, C. Terragna, S. Durante, E. Valli, T. Kalebic, R. Bernardoni, C. Perrod, M. Haber, M.D. Norri, M. Baccarani, G. Martinelli, and G. Perini
- Subjects
Cancer Research ,Transcription, Genetic ,Abcg2 ,ABCG2 ,Population ,Antigens, CD34 ,ATP-binding cassette transporter ,CD34+ PROGENITOR CELLS ,Proto-Oncogene Proteins c-myc ,ABC TRANSPORTERS ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,Transcriptional regulation ,medicine ,C-MYC ,ATP Binding Cassette Transporter, Subfamily G, Member 2 ,Humans ,Gene Regulatory Networks ,Epigenetics ,Promoter Regions, Genetic ,education ,Molecular Biology ,Cells, Cultured ,Cell Proliferation ,education.field_of_study ,biology ,Cytotoxins ,Myeloid leukemia ,DNA Methylation ,Hematopoietic Stem Cells ,medicine.disease ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Oncology ,Drug Resistance, Neoplasm ,biology.protein ,Cancer research ,ATP-Binding Cassette Transporters ,CpG Islands ,Efflux ,CHRONIC MYELOGENOUS LEUKAEMIA ,Chronic myelogenous leukemia - Abstract
Resistance to chemotherapeutic agents remains one of the major impediments to a successful treatment of chronic myeloid leukemia (CML). Misregulation of the activity of a specific group of ATP-binding cassette transporters (ABC) is responsible for reducing the intracellular concentration of drugs in leukemic cells. Moreover, a consistent body of evidence also suggests that ABC transporters play a role in cancer progression beyond the efflux of cytotoxic drugs. Despite a large number of studies that investigated the function of the ABC transporters, little is known about the transcriptional regulation of the ABC genes. Here, we present data showing that the oncoprotein c-MYC is a direct transcriptional regulator of a large set of ABC transporters in CML. Furthermore, molecular analysis carried out in CD34+ hematopoietic cell precursors of 21 CML patients reveals that the overexpression of ABC transporters driven by c-MYC is a peculiar characteristic of the CD34+ population in CML and was not found either in the population of mononuclear cells from which they had been purified nor in CD34+ cells isolated from healthy donors. Finally, we describe how the methylation state of CpG islands may regulate the access of c-MYC to ABCG2 gene promoter, a well-studied gene associated with multidrug resistance in CML, hence, affecting its expression. Taken together, our findings support a model in which c-MYC–driven transcriptional events, combined with epigenetic mechanisms, direct and regulate the expression of ABC genes with possible implications in tumor malignancy and drug efflux in CML. Mol Cancer Res; 9(8); 1054–66. ©2011 AACR.
- Published
- 2011
36. Mutations in the 5' UTR of ANKRD26, the ankirin repeat domain 26 gene, cause an autosomal-dominant form of inherited thrombocytopenia, THC2
- Author
-
Mariateresa Di Stazio, Marcella Ferraro, Carlo L. Balduini, Pamela Magini, Patrizia Noris, Francesca Punzo, Chiara Gnan, Saverio Scianguetta, Luca Dezzani, Samuele Gherardi, Daniela De Rocco, Tommaso Pippucci, Caterina Marconi, Valeria Bozzi, Giuseppe Loffredo, Nuria Pujol-Moix, Serena Barozzi, Giovanni Castegnaro, Anna Savoia, Giovanni Perini, Marco Seri, Silverio Perrotta, Alessandro Pecci, Pippucci T, Savoia A, Perrotta S, Pujol-Moix N, Noris P, Castegnaro G, Pecci A, Gnan C, Punzo F, Marconi C, Gherardi S, Loffredo G, De Rocco D, Scianguetta S, Barozzi S, Magini P, Bozzi V, Dezzani L, Di Stazio M, Ferraro M, Perini G, Seri M, Balduini CL, Pippucci, T, Savoia, A, Perrotta, Silverio, Pujol Moix, N, Noris, P, Castegnaro, G, Pecci, A, Gnan, C, Punzo, F, Marconi, C, Gherardi, S, Loffredo, G, De Rocco, D, Scianguetta, S, Barozzi, S, Magini, P, Bozzi, V, Dezzani, L, Di Stazio, M, Ferraro, M, Perini, G, Seri, M, Balduini, C. L., Pippucci, Tommaso, Savoia, Anna, Pujol Moix, Núria, Noris, Patrizia, Castegnaro, Giovanni, Pecci, Alessandro, Gnan, Chiara, Punzo, Francesca, Marconi, Caterina, Gherardi, Samuele, Loffredo, Giuseppe, DE ROCCO, Daniela, Scianguetta, Saverio, Barozzi, Serena, Magini, Pamela, Bozzi, Valeria, Dezzani, Luca, DI STAZIO, Mariateresa, Ferraro, Marcella, Perini, Giovanni, Seri, Marco, and Balduini, Carlo L.
- Subjects
Untranslated region ,Male ,Five prime untranslated region ,Molecular Sequence Data ,Locus (genetics) ,Chromosome Disorders ,autosomal-dominant thrombocytopenia ,Haploinsufficiency ,Biology ,Genome ,Conserved sequence ,Genetic ,Ankyrin Repeat ,Base Sequence ,Chromosome Breakage ,Conserved Sequence ,Female ,Genetic Loci ,Humans ,Pedigree ,Thrombocytopenia ,Genes, Dominant ,Mutation ,Genetics ,Genetics (clinical) ,Report ,Genetics(clinical) ,Dominant ,Gene ,ANKRD26 ,Molecular biology ,Chromosome Disorder ,Genes ,THC2 ,Chromosome breakage ,Human - Abstract
THC2, an autosomal-dominant thrombocytopenia described so far in only two families, has been ascribed to mutations in MASTL or ACBD5. Here, we show that ANKRD26, another gene within the THC2 locus, and neither MASTL nor ACBD5, is mutated in eight unrelated families. ANKRD26 was also found to be mutated in the family previously reported to have an ACBD5 mutation. We identified six different ANKRD26 mutations, which were clustered in a highly conserved 19 bp sequence located in the 5' untranslated region. Mutations were not detected in 500 controls and are absent from the 1000 Genomes database. Available data from an animal model and Dr. Watson's genome give evidence against haploinsufficiency as the pathogenetic mechanism for ANKRD26-mediated thrombocytopenia. The luciferase reporter assay suggests that these 5' UTR mutations might enhance ANKRD26 expression. ANKRD26 is the ancestor of a family of primate-specific genes termed POTE, which have been recently identified as a family of proapoptotic proteins. Dysregulation of apoptosis might therefore be the pathogenetic mechanism, as demonstrated for another thrombocytopenia, THC4. Further investigation is needed to provide evidence supporting this hypothesis.
- Published
- 2011
37. Transcriptional upregulation of histone deacetylase 2 promotes Myc-induced oncogenic effects
- Author
-
Eric Sekyere, Michelle Haber, Zillan Neiron, Christopher J. Scarlett, Fabio Stossi, David K. Chang, Ning Xu, Jankowski K, Nunzio Iraci, Glenn M. Marshall, Benita S. Katzenellenbogen, Tao Liu, Georg von Jonquieres, Samuele Gherardi, Giovanni Perini, Pei Yan Liu, Andrew V. Biankin, Toby Trahair, Murray D. Norris, J. Keating, Marshall G.M., Gherardi S., Xu N., Neiron Z., Trahair T., Scarlett C.J., Chang D.K., Liu P.Y., Jankowski K., Iraci N., Haber M., Norris M.D., Keating J., Sekyere E., Jonquieres G., Stossi F., Katzenellenbogen B.S., Biankin AV, Perini G., and Liu T.
- Subjects
Chromatin Immunoprecipitation ,Cancer Research ,Transcription, Genetic ,Cyclin G2 ,Histone Deacetylase 2 ,Mice, Transgenic ,medicine.disease_cause ,Proto-Oncogene Proteins c-myc ,Mice ,Downregulation and upregulation ,Cell Line, Tumor ,MYCN ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Cell Proliferation ,DNA Primers ,CCGN2 ,Base Sequence ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Histone deacetylase 2 ,Cell growth ,HDAC PROTEINS ,Cell cycle ,PANCREATIC CANCER ,Up-Regulation ,Pancreatic Neoplasms ,Histone ,NEUROBLASTOMA ,biology.protein ,Cancer research ,Histone deacetylase ,Carcinogenesis ,Chromatin immunoprecipitation - Abstract
Myc oncoproteins and histone deacetylases (HDACs) modulate gene transcription and enhance cancer cell proliferation, and HDAC inhibitors are among the most promising new classes of anticancer drugs. Here, we show that N-Myc and c-Myc upregulated HDAC2 gene expression in neuroblastoma and pancreatic cancer cells, respectively, which contributed to N-Myc- and c-Myc-induced cell proliferation. Cyclin G2 (CCNG2) was commonly repressed by N-Myc and HDAC2 in neuroblastoma cells and by c-Myc and HDAC2 in pancreatic cancer cells, and could be reactivated by HDAC inhibitors. 5-bromo-2'-deoxyuridine incorporation assays showed that transcriptional repression of CCNG2 was, in part, responsible for N-Myc-, c-Myc- and HDAC2-induced cell proliferation. Dual crosslinking chromatin immunoprecipitation assay demonstrated that N-Myc acted as a transrepressor by recruiting the HDAC2 protein to Sp1-binding sites at the CCNG2 gene core promoter. Moreover, HDAC2 was upregulated, and CCNG2 downregulated, in pre-cancerous and neuroblastoma tissues from N-Myc transgenic mice, and c-Myc overexpression correlated with upregulation of HDAC2 and repression of CCNG2 in tumour tissues from pancreatic cancer patients. Taken together, our data indicate the critical roles of upregulation of HDAC2 and suppression of CCNG2 in Myc-induced oncogenesis, and have significant implications for the application of HDAC inhibitors in the prevention and treatment of Myc-driven cancers.
- Published
- 2010
38. p53 Is a Direct Transcriptional Target of MYCN in Neuroblastoma
- Author
-
John Lunec, Katrina M Wood, Deborah A. Tweddle, Giovanni Perini, Lindi Chen, Laura D. Gamble, Nunzio Iraci, Samuele Gherardi, Chen L., Iraci N., Gherardi S., Gamble L.D., Wood K.M., Perini G., Lunec J., and Tweddle D.A.
- Subjects
Cancer Research ,Tumor suppressor gene ,Transcription, Genetic ,Apoptosis ,Biology ,Transfection ,N-Myc Proto-Oncogene Protein ,Article ,CHROMATIN IMMUNOPRECIPITATION ,Neuroblastoma ,MYCN ,medicine ,Tumor Cells, Cultured ,Humans ,TRANSCRIPTION ,RNA, Small Interfering ,Promoter Regions, Genetic ,neoplasms ,Oligonucleotide Array Sequence Analysis ,Regulation of gene expression ,Oncogene Proteins ,Gene knockdown ,P53 ,Brain Neoplasms ,Gene Expression Profiling ,Gene Amplification ,Nuclear Proteins ,medicine.disease ,Genes, p53 ,Molecular biology ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,Oncology ,NEUROBLASTOMA ,Cancer research ,biology.protein ,Mdm2 ,Tumor Suppressor Protein p53 ,Chromatin immunoprecipitation - Abstract
MYCN amplification occurs in around 25% of neuroblastomas, and is associated with rapid tumor progression and poor prognosis. MYCN plays a paradoxical role in driving cellular proliferation and inducing apoptosis. We previously observed nuclear p53 accumulation in neuroblastoma and hypothesize that MYCN regulates p53 in neuroblastoma. Immunohistochemical analysis of 82 neuroblastoma tumors demonstrated an association between high p53 expression and MYCN expression and MYCN amplification. In a panel of 5 MYCN amplified and 5 non-amplified neuroblastoma cell lines and also the Tet21N regulatable MYCN expression system there was a correlation between p53 expression and MYCN expression. Knockdown of MYCN in 2 MYCN amplified cell lines led to a decrease in p53 expression. Tet21N MYCN+ cells expressed higher p53 mRNA and protein, and had greater p53 transcriptional activity, in comparison with Tet21N MYCN− cells. Using chromatin immunoprecipitation and reporter gene assays, MYCN was found to bind directly to an E-Box motif located close to the transcriptional start site within the p53 promoter and initiate transcription. Mutation of the E-Box led to a decrease in MYCN driven transcriptional activity. Microarray analysis of Tet21N MYCN+/− cells showed that several p53 regulated genes were upregulated in the presence of MYCN, including MDM2 and PUMA. Knockdown of MYCN and p53 in a MYCN amplified cell line led to reduced PUMA levels and other markers of apoptosis. We conclude that MYCN transcriptionally upregulates p53 expression in neuroblastoma and may be an important mechanism by which MYCN induces apoptosis.
- Published
- 2010
39. Direct and coordinate regulation of ATP-binding cassette transporter genes by Myc factors generates specific transcription signatures that significantly affect the chemoresistance phenotype of cancer cells
- Author
-
Glenn M. Marshall, Daniel Diolaiti, Michelle J. Henderson, Claudia Flemming, Giovanni Perini, Jason W. H. Wong, Murray D. Norris, Michelle Haber, Marcia A. Munoz, Manfred Schwab, Antonio Porro, Samuele Gherardi, Emanuele Valli, Chengyuan Xue, Giuliano Della Valle, Nunzio Iraci, Porro A., Haber M., Diolaiti D., Iraci N., Henderson M., Gherardi S., Valli E., Munoz M.A., Xue C., Flemming C., Schwab M., Wong J.H., Marshall G.M., Della Valle G., Norris M.D., and Perini G.
- Subjects
Transcription, Genetic ,Repressor ,ATP-binding cassette transporter ,Biology ,Biochemistry ,ABC DRUG TRANSPORTERS ,Proto-Oncogene Proteins c-myc ,Inhibitory Concentration 50 ,CHROMATIN IMMUNOPRECIPITATION ,Cell Line, Tumor ,Neoplasms ,Gene expression ,MYCN ,Transcriptional regulation ,Humans ,Gene Regulation ,Molecular Biology ,Genetics ,Models, Genetic ,Activator (genetics) ,Brain Neoplasms ,Gene Expression Profiling ,Retinoblastoma ,ABCB5 ,Cell Biology ,CANCER ,Cell biology ,Gene expression profiling ,Phenotype ,Drug Resistance, Neoplasm ,NEUROBLASTOMA ,ATP-Binding Cassette Transporters ,Drug Screening Assays, Antitumor ,Chromatin immunoprecipitation ,Transcription Factors - Abstract
Increased expression of specific ATP-binding cassette (ABC) transporters is known to mediate the efflux of chemotherapeutic agents from cancer cells. Therefore, establishing how ABC transporter genes are controlled at their transcription level may help provide insight into the role of these multifaceted transporters in the malignant phenotype. We have investigated ABC transporter gene expression in a large neuroblastoma data set of 251 tumor samples. Clustering analysis demonstrated a strong association between differential ABC gene expression patterns in tumor samples and amplification of the MYCN oncogene, suggesting a correlation with MYCN function. Using expression profiling and chromatin immunoprecipitation studies, we show that MYCN oncoprotein coordinately regulates transcription of specific ABC transporter genes, by acting as either an activator or a repressor. Finally, we extend these notions to c-MYC showing that it can also regulate the same set of ABC transporter genes in other tumor cells through similar dynamics. Overall our findings provide insight into MYC-driven molecular mechanisms that contribute to coordinate transcriptional regulation of a large set of ABC transporter genes, thus affecting global drug efflux.
- Published
- 2010
40. Abstract 789: GSTP1 expression is regulated by MycN and is a marker of poor outcome in childhood neuroblastoma
- Author
-
Murray D. Norris, Glenn M. Marshall, Catherine Burkhart, Amanda J. Russell, Jamie I. Fletcher, Michelle Haber, Wendy B. London, Lesley J. Ashton, André Oberthuer, Emanuele Valli, Samuele Gherardi, Jayne Murray, and Giovanni Perini
- Subjects
Cancer Research ,Microarray ,Cancer ,Biology ,medicine.disease ,Phenotype ,Molecular biology ,GSTP1 ,Oncology ,Neuroblastoma ,medicine ,Cancer research ,Transcriptional regulation ,Childhood Neuroblastoma ,neoplasms ,Chromatin immunoprecipitation - Abstract
In childhood neuroblastoma, amplification of the transcription factor MycN is associated with poor outcome and a multidrug resistant phenotype. We have previously shown that MycN regulates the expression of several ATP-binding cassette (ABC) transporter genes, and that these genes represent powerful prognostic markers in neuroblastoma (1,2,3). Although these transporters are able to efflux a wide range of cytotoxic drugs, they do not confer resistance to several important cytotoxic agents used to treat neuroblastoma, including cisplatin and cyclophosphamide, hence we explored the prognostic significance and transcriptional regulation of the phase II detoxifying enzyme, glutathione S-transferase P1 (GSTP1). Using quantitative real-time PCR, GSTP1 gene expression was assessed in a retrospective cohort of 51 patients, and subsequently in a cohort of 207 prospectively accrued primary neuroblastomas. These data, and GSTP1 expression data from an independent microarray study of 251 neuroblastoma samples, were correlated with established prognostic indicators and disease outcome. High levels of GSTP1 were associated with decreased event-free and overall survival in all three cohorts. Multivariate analyses, including age at diagnosis, tumor stage and MYCN amplification status, were conducted on the two larger cohorts and demonstrated independent prognostic significance of GSTP1 expression levels. We also explored the regulation of GSTP1 by MycN using chromatin immunoprecipitation (ChIP) and luciferase reporter assays in neuroblastoma cell lines and found that GSTP1 is a direct transcriptional target of MycN. We conclude that N-Myc regulates multiple components of drug metabolism and efflux pathways, including GSTP1 and transporters of the ABC superfamily. The regulation of these key multidrug resistance mechanisms may have particular significance for malignancies with amplification of Myc family genes. 1) J Biol Chem, 285:19532-19543, 2010 2) J Natl Cancer Institute, 103:1236-51, 2011 4) Nat Rev Cancer, 10:147-156, 2010 Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 789. doi:1538-7445.AM2012-789
- Published
- 2012
- Full Text
- View/download PDF
41. SIRT1 Promotes N-Myc Oncogenesis through a Positive Feedback Loop Involving the Effects of MKP3 and ERK on N-Myc Protein Stability
- Author
-
Toby Trahair, Ning Xu, Andrew V. Biankin, Eric Sekyere, Murray D. Norris, Wayne Thomas, Michelle Haber, Emanuele Valli, Antonio Bedalov, Christopher J. Scarlett, Margo van Bekkum, Dora Ling, Glenn M. Marshall, Pei Y. Liu, Kacper Jankowski, Giovanni Perini, Samuele Gherardi, Tao Liu, Nuncio Iraci, Karen L. MacKenzie, G.M. Marshall, P.Y. Liu, S. Gherardi, C.J. Scarlett, A. Bedalov, N. Xu, N. Iraci, E. Valli, D. Ling, W. Thoma, M. van Bekkum, E. Sekyere, K. Jankowski, T. Trahair, K.L. Mackenzie, M. Haber, M.D. Norri, A.V. Biankin, G. Perini, and T. Liu.
- Subjects
Cancer Research ,MPK3 ,environment and public health ,STABILITA' PROTEICA ,Mice ,Random Allocation ,Sirtuin 1 ,MYCN ,Protein phosphorylation ,Enzyme Inhibitors ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Promoter Regions, Genetic ,SIRT1 ,NEUROBLASTOMA ,Neurological Tumors ,Genetics (clinical) ,Feedback, Physiological ,Protein Stability ,Autophagy-related protein 13 ,Tumor Burden ,Gene Expression Regulation, Neoplastic ,Oncology ,GATAD2B ,Medicine ,Protein stabilization ,Research Article ,lcsh:QH426-470 ,Sp1 Transcription Factor ,DNA transcription ,Mice, Transgenic ,Pyrimidinones ,Naphthalenes ,Biology ,Proto-Oncogene Proteins c-myc ,Retinoblastoma-like protein 1 ,Dual Specificity Phosphatase 6 ,Cell Line, Tumor ,Genetics ,Animals ,Protein kinase A ,Carbohydrate-responsive element-binding protein ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Cell Proliferation ,Binding Sites ,YY1 ,Cancers and Neoplasms ,lcsh:Genetics ,enzymes and coenzymes (carbohydrates) ,Cancer research ,Gene expression - Abstract
The N-Myc oncoprotein is a critical factor in neuroblastoma tumorigenesis which requires additional mechanisms converting a low-level to a high-level N-Myc expression. N-Myc protein is stabilized when phosphorylated at Serine 62 by phosphorylated ERK protein. Here we describe a novel positive feedback loop whereby N-Myc directly induced the transcription of the class III histone deacetylase SIRT1, which in turn increased N-Myc protein stability. SIRT1 binds to Myc Box I domain of N-Myc protein to form a novel transcriptional repressor complex at gene promoter of mitogen-activated protein kinase phosphatase 3 (MKP3), leading to transcriptional repression of MKP3, ERK protein phosphorylation, N-Myc protein phosphorylation at Serine 62, and N-Myc protein stabilization. Importantly, SIRT1 was up-regulated, MKP3 down-regulated, in pre-cancerous cells, and preventative treatment with the SIRT1 inhibitor Cambinol reduced tumorigenesis in TH-MYCN transgenic mice. Our data demonstrate the important roles of SIRT1 in N-Myc oncogenesis and SIRT1 inhibitors in the prevention and therapy of N-Myc–induced neuroblastoma., Author Summary The class III histone deacetylase SIRT1 is repressed by tumor suppressor genes and exerts divergent effects on tumorigenesis depending on its down-stream targets. Small molecule SIRT1 inhibitors have shown promising anti-cancer effects both in vitro and in vivo. Here we identified SIRT1 as a gene directly up-regulated by N-Myc and identified SIRT1-mediated transcriptional repression as a novel mechanism responsible for maintaining N-Myc oncoprotein stability. Moreover, SIRT1 contributed to N-Myc–induced cell proliferation, and preventative treatment with the SIRT1 inhibitor Cambinol reduced tumorigenesis in N-Myc transgenic mice. Our data identify SIRT1 as an important co-factor for N-Myc oncogenesis and provide important evidence for the potential application of SIRT1 inhibitors in the prevention and therapy of N-Myc–induced neuroblastoma.
- Published
- 2011
- Full Text
- View/download PDF
42. Abstract 3027: SIRT1 promotes N-Myc oncogenesis stabilizing N-Myc protein
- Author
-
Ning Xu, Tao Liu, Andrew V. Biankin, Pei Y. Liu, Michelle Haber, Samuele Gherardi, Murray D. Norris, Glenn M. Marshall, Giovanni Perini, Christopher J. Scarlett, Nunzio Iraci, Antonio Bedalov, and Karen L. MacKenzie
- Subjects
MAPK/ERK pathway ,Cancer Research ,Oncology ,Transcriptional repressor complex ,Cancer research ,Phosphorylation ,Protein phosphorylation ,Histone deacetylase ,Biology ,Protein stabilization ,Protein kinase A ,N-Myc - Abstract
The N-Myc oncoprotein is a critical factor in neuroblastoma tumorigenesis which requires additional mechanisms converting low-level to high-level N-Myc expression. N-Myc protein is stabilized when phosphorylated at Serine 62 by phosphorylated ERK protein. Here we describe a novel positive feedback loop whereby N-Myc directly induced the transcription of the histone deacetylase SIRT1, which in turn increased N-Myc protein stability. SIRT1 binds to Myc Box I domain of N-Myc protein to form a novel transcriptional repressor complex at gene promoter of mitogen-activated protein kinase phosphatase 3 (MKP3), leading to transcriptional repression of MKP3, ERK protein phosphorylation, N-Myc protein phosphorylation at Serine 62 and N-Myc protein stabilization. Importantly, SIRT1 was up-regulated, MKP3 down-regulated, in pre-cancerous cells, and preventative treatment with the SIRT1 inhibitor, Cambinol, reduced tumorigenesis in TH-MYCN transgenic mice. Our data demonstrate the important roles of SIRT1 in N-Myc oncogenesis and SIRT1 inhibitors in the prevention and therapy of N-Myc-induced neuroblastoma. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 3027. doi:10.1158/1538-7445.AM2011-3027
- Published
- 2011
- Full Text
- View/download PDF
43. Abstract 954: ABCC/MRP multidrug transporters contribute to neuroblastoma biology, pathogenesis and clinical outcome, independently of any role in cytotoxic drug efflux
- Author
-
Allen Buxton, Wendy B. London, Murray D. Norris, Jayne Murray, Lesley J. Ashton, Jamie I. Fletcher, Michelle J. Henderson, Michelle Haber, Glenn M. Marshall, Antonio Porro, Amanda J. Russell, Samuele Gherardi, Marcia A. Munoz, Alan C. Sartorelli, Giovanni Perini, Claudia Flemming, Susan L. Cohn, Alan Kwek, Manfred Schwab, Chengyuan Xue, and Nunzio Iraci
- Subjects
Pathogenesis ,Cancer Research ,Oncology ,Cytotoxic drug ,Neuroblastoma ,medicine ,Efflux ,Biology ,Pharmacology ,medicine.disease ,Multidrug transporter - Abstract
We have previously shown that high levels of the multidrug transporters ABCC1/MRP1 (1,2) and ABCC4/MRP4 (3) are strongly predictive of poor outcome in the childhood cancer, neuroblastoma. Although the prognostic significance of ABCC1 may be explained in terms of cytotoxic drug resistance, none of the drugs used to treat children in these studies were ABCC4 substrates. This suggests that multidrug transporters can contribute to the malignant phenotype, independent of cytotoxic drug efflux, as we have recently outlined (4). To address this hypothesis, a MYCN-driven transgenic mouse neuroblastoma model was crossed with an Abcc1-deficient mouse strain or alternatively, treated with an ABCC1 inhibitor. Pharmacological inhibition or genetic depletion of ABCC1 significantly inhibited neuroblastoma development in MYCN transgenic mice, while knockdown of ABCC1 using siRNA reduced cell motility and clonogenicity in cultured neuroblastoma cells, and induced morphological differentiation. Analysis of a large neuroblastoma cohort of 209 primary untreated tumors revealed that amongst the 12 members of the ABCC gene family, expression of only ABCC1, ABCC4 and also ABCC3 were predictive of neuroblastoma outcome. We confirmed a highly significant association between high levels of either ABCC1 or ABCC4 and poor outcome (p 1) New Engl J Med 334:231-238, 1996 2) J Clin Oncol 24:1546-53, 2006 3) Mol Cancer Ther 4:547-53, 2005 4) Nat Rev Cancer, 10:147-156, 2010 Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 954. doi:10.1158/1538-7445.AM2011-954
- Published
- 2011
- Full Text
- View/download PDF
44. Abstract 4869: Transcriptional upregulation of histone deacetylase 2 promotes Myc-induced oncogenic effects
- Author
-
Benita S. Katzenellenbogen, Samuele Gherardi, Pei Y. Liu, Andrew V. Biankin, Kacper Jankowski, Giovanni Perini, Toby Trahair, Tao Liu, Fabio Stossi, Murray D. Norris, David K. Chang, Nunzio Iraci, Christopher J. Scarlett, Michelle Haber, Ning Xu, Glenn M. Marshall, and Zillan Neiron
- Subjects
Cancer Research ,biology ,Histone deacetylase 2 ,Cell growth ,Promoter ,medicine.disease_cause ,medicine.disease ,Molecular biology ,Histone ,Oncology ,Pancreatic cancer ,Neuroblastoma ,biology.protein ,medicine ,Cancer research ,Carcinogenesis ,Chromatin immunoprecipitation - Abstract
Myc oncoproteins induce cancer cell proliferation by transcriptional modulation. Histone deacetylases (HDACs) enhance cancer cell proliferation and survival, in part, by effects on tumour suppressor gene transcription, and thus HDAC inhibitors are among the most promising new classes of anticancer drugs. Here we show that N-Myc and c-Myc up-regulated HDAC2 gene expression in neuroblastoma and pancreatic cancer cells respectively, which contributed to N-Myc- and c-Myc-induced cell proliferation. Affymetrix microarray revealed that a subset of genes, including cyclin G2 (CCNG2), was commonly repressed by N-Myc and HDAC2 in neuroblastoma cells. PCR analysis confirmed that CCNG2 gene transcription was commonly repressed by N-Myc and HDAC2 in neuroblastoma cells and by c-Myc and HDAC2 in pancreatic cancer cells, and could be reactivated by HDAC inhibitors. BrdU incorporation assays demonstrated that transcriptional repression of CCNG2 was, in part, responsible for N-Myc-, c-Myc and HDAC2-induced cell proliferation. Dual cross-linking chromatin immunoprecipitation and protein co-immunoprecipitation assays showed that N-Myc acted as a transrepressor, by recruiting the HDAC2 protein to an Sp1-binding site at the CCNG2 core promoter, in a manner distinct from its action as a transactivator. Moreover, HDAC2 was up-regulated, and CCNG2 down-regulated, in pre-cancerous and neuroblastoma tissues from MYCN (N-Myc) transgenic mice, and c-Myc over-expression correlated with up-regulation of HDAC2 and repression of CCNG2 in tumour tissues from pancreatic cancer patients. Taken together, our data indicate the critical roles of up-regulation of HDAC2 and suppression of CCNG2 in Myc-induced oncogenesis, and have significant implications for the application of HDAC inhibitors in the prevention and treatment of Myc-driven cancers. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4869.
- Published
- 2010
- Full Text
- View/download PDF
45. C-Myc Mediated Regulation of Multidrug Resistance Genes in Chronic Myeloid Leukaemia Cd34+ Cell Progenitors
- Author
-
Nunzio Iraci, Thea Kalebic, Sandra Durante, Annalisa Astolfi, Carolina Terragna, Giuseppe Saglio, Simona Soverini, Emanuele Valli, Fabrizio Pane, Giovanni Perini, Giovanni Martinelli, Michele Baccarani, Samuele Gherardi, and Roberto Bernardoni
- Subjects
education.field_of_study ,biology ,Immunology ,Population ,ATP-binding cassette transporter ,Promoter ,Cell Biology ,Hematology ,ABCC4 ,Biochemistry ,Molecular biology ,Imatinib mesylate ,hemic and lymphatic diseases ,Gene expression ,biology.protein ,Gene silencing ,education ,Chromatin immunoprecipitation - Abstract
Abstract 3252 Poster Board III-1 A better understanding of the mechanisms which regulate drug resistance is critical for improving therapy for patients at risk of poor response. Even for CML patients the long-term benefits of the treatment are limited by the emergence of resistance, although the therapy has been dramatically improved by Imatinib Mesylate, which inhibits Bcr-Abl activity. One of the most important mechanisms of resistance is the disregulation of various members of the highly conserved family of transmembrane proteins characterized by an ATP-binding cassette domain, called ABC superfamily of transporters (Dean et al., Genome Res 2001). In CML cells an aberrant expression of ABC transporter genes has been described to be mediated by specific transcription factors, which are in turn affected by an aberrant activity of Bcr-Abl. It has been shown that Bcr-Abl can indirectly activate c-Myc via the JAK2 pathway, which increases a translation of c-Myc mRNA. In addition, in a subgroup of CML patients a chromosome 8 trysomy, has been observed, which can be associated with c-Myc amplification. In this study we have investigated whether c-Myc can regulate transcription of ABC tranporter genes in CML. Initially, ABC transporter gene expression has been monitored in HL60, a human promyelocytic cell line in which c-MYC is overexpressed. We have examined the expression level of all 48 human ABCs transporters as a function of c-MYC silencing. Our results have demonstrated that c-Myc regulates the transcription of several ABC genes, such as ABCA2, ABCB9, ABCB10, ABCC1, ABCC4, ABCE1, ABCF1, ABCF2, a majority of which has been found implicated in drug resistance. Furthermore, by performing chromatin immunoprecipitation (ChIP) we have shown a direct binding of c-Myc to the promoters of those ABC transporter genes in HL60 cell line. In addition, by ChIP we have demonstrated that c-MYC is physically bound to the promoter of tested ABC genes in CML cell lines KG-1a and K562. Based on those findings we have investigated the expression level of c-Myc in CD34+ progenitor cells derived from CML patients. Also, we have evaluated the effects of highly expressed c-Myc on ABC transporters. Our results have shown in a group of 20 newly diagnosed chronic phase (CP)-CML patients an increased expression of c-Myc in CD34+ cell fraction, when compared to the expression level of c-Myc in the entire population of mononuclear cells from which CD34+ cell fraction has been purified. In those cells we have identified in association with an increased level of c-Myc an increased expression of the same subset of ABC genes, which we have observed in cell lines. Furthermore, we have evaluated whether a differential expression of c-Myc and ABC transporter genes is associated with low Sokal risk and high Sokal risk patients. This analysis has been performed on CD34+ cells purified from 19 CML patients of whom 12 were scored as low Sokal risk and 7 as high Sokal risk. Our results have shown that expression of c-Myc and ABCC4 is different in those two patients population (p Disclosures: No relevant conflicts of interest to declare.
- Published
- 2009
- Full Text
- View/download PDF
46. Suppression of Bcr-Abl Expression in CML by A Panel of miRNAs
- Author
-
Nunzio Iraci, Thea Kalebic, Samuele Gherardi, Michele Baccarani, Simona Soverini, Giovanni Martinelli, Emanuele Valli, and Giovanni Perini
- Subjects
Untranslated region ,Immunology ,HEK 293 cells ,Translation (biology) ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Molecular biology ,MiRBase ,Cell biology ,Imatinib mesylate ,hemic and lymphatic diseases ,microRNA ,Gene silencing ,Gene - Abstract
Abstract 854 It has been proposed that most protein-encoding genes may be regulated by small multifunctional RNAs which can control transcript turnover and/or protein translation (siRNA and miRNA). Specifically, miRNAs have been shown to act predominantly at the level of translation by blocking the access or sliding of ribosomes to mRNAs (3'UTR). Several studies have shown that multiple miRNAs can be disregulated in tumours as compared to normal tissues. For example, the miR-17-92 cluster is up regulated in CML, suggesting their involvement in leukemiagenesis (Venturini et al., Blood 2007). However, a role of miRNA in preventing tumor development and progression has also been suggested. Bueno et al have shown that hsa-miR-203 can specifically target BCR-ABL and reduce its expression in CML derived cell lines (Bueno et al., Cancer Cell 2008). Since multiple miRNAs seem to act in a combinatorial fashion to regulate mRNA translation, we hypothesised that other miRNAs in addition to hsa-miR-203 might be involved in BCR-ABL expression control. To test this hypothesis we conducted a search for miRNAs specifically targeting Bcr-Abl, using the miRBASE program to scan human genome (http://microrna.sanger.ac.uk/). This search identified 15 miRNAs potentially able to target the BCR-ABL 3' UTR. Further investigation showed that only hsa-miR-451, hsa-miR-515-3p and hsa-miR-760 had a sufficiently high score to predict genuine interactions with the 3'UTR of BCR-ABL and therefore only these miRNAs were utilized for further analyses. Initially, the three miRNAs were transfected in K562 Ph+ cells, together with hsa-miR-203 as a positive control and a scrambled miRNA used as a negative control. BCR-ABL expression was monitored at both mRNA ( qRT-PCR) and protein level (Western Blotting). Our results demonstrated that BCR-ABL mRNA expression was unaffected by miRNAs, whereas protein expression was significantly reduced. Considering the combinatorial function of miRNAs on their target mRNAs, we also tested whether a pool of the four identified miRNAs could be effective in suppressing BCR-ABL expression. In those experiments, the molar concentration of each miRNA was a quarter of that used for a single miRNA transfection. The results have shown that the pool of miRNAs worked efficiently in suppressing Bcr-Abl expression, which suggested a cooperative function of the four miRNAs in controlling both the expression and translation of Bcr-Abl. To further confirm that miRNAs directly targeted BCR-ABL, the 3'UTR of the gene was cloned downstream of a reporter renilla luciferase gene. The reporter was co-transfected in HEK293 cells together with single miRNAs or a pool of them, and luciferase activity was quantified. Those results show that the presence of each miRNAs significantly reduced the luciferase activity as compared to that obtained by transfecting cells with a scrambled miRNA. These experiments therefore confirmed a direct effect of the four miRNAs on the 3'UTR of BCR-ABL. Again the “pool” of miRNAs showed the strongest effect on the luc reporter expression. Through an additional deletion analysis we mapped the regions of 3'UTR of BCR-ABL targeted by the four miRNAs, in order to confirm that the predicted binding regions of miRNAs are critical to mediate repression of BCR-ABL. In conclusion, our study identified three new human miRNAs having a potential to specifically target BCR-ABL and suppress its translation and expression in CML cells. BCR-ABL, which plays a critical role in CML, is effectively suppressed by TK inhibitors, exemplified by Gleevec and our findings provide a rationale to exploit miRNA as an alternative therapeutic approache which could further improve CML treatment, or to complement TK inhibitors in an effort to eradicate minimal residual disease. Supported by: Novartis Oncology, Clinical Development, TOPS Clinical Correlative Studies Network Disclosures: No relevant conflicts of interest to declare.
- Published
- 2009
- Full Text
- View/download PDF
47. MYCN amplification confers enhanced folate dependence and methotrexate sensitivity in neuroblastoma
- Author
-
Murray D. Norris, André Oberthuer, Giovanni Perini, Lesley J. Ashton, Glenn M. Marshall, Benedikt Brors, Chengyuan Xue, Dilafruz Juraeva, Michelle Haber, Jamie I. Fletcher, Claudia Flemming, Samuele Gherardi, Matthias Fischer, Diana T. Lau, Lau, Diana T, Flemming, Claudia L, Gherardi, Samuele, Perini, Giovanni, Oberthuer, André, Fischer, Matthia, Juraeva, Dilafruz, Brors, Benedikt, Xue, Chengyuan, Norris, Murray D, Marshall, Glenn M, Haber, Michelle, Fletcher, Jamie I, and Ashton, Lesley J
- Subjects
medicine.medical_specialty ,MYC ,Biology ,N-Myc Proto-Oncogene Protein ,methotrexate ,chemistry.chemical_compound ,Neuroblastoma ,Reduced Folate Carrier Protein ,Folic Acid ,Internal medicine ,Cell Line, Tumor ,MYCN ,medicine ,Humans ,RNA, Messenger ,neoplasms ,Oncogene Proteins ,Hematology ,Gene Amplification ,Intracellular Signaling Peptides and Proteins ,Cancer ,Nuclear Proteins ,medicine.disease ,Molecular medicine ,Primary tumor ,3. Good health ,Gene Expression Regulation, Neoplastic ,Oncology ,chemistry ,Antifolate ,Cancer research ,SLC19A1 ,Folic Acid Antagonists ,Methotrexate ,medicine.drug ,Research Paper - Abstract
// Diana T. Lau 1 , Claudia L. Flemming 1 , Samuele Gherardi 2 , Giovanni Perini 2 , Andre Oberthuer 3 , Matthias Fischer 3 , Dilafruz Juraeva 4 , Benedikt Brors 4 , Chengyuan Xue 1 , Murray D. Norris 1 , Glenn M. Marshall 1,5 , Michelle Haber 1 , Jamie I. Fletcher 1,* and Lesley J. Ashton 6,* 1 Children’s Cancer Institute Australia, Lowy Cancer Research Centre, Randwick, NSW, Australia 2 Department of Biology, University of Bologna, Bologna, Italy 3 Children’s Hospital, Department of Pediatric Oncology and Hematology, University of Cologne and Centre for Molecular Medicine Cologne, Cologne, Germany 4 Division of Theoretical Bioinformatics, German Cancer Research Center, Heidelberg, Germany 5 Kids Cancer Centre, Sydney Children’s Hospital, Randwick, NSW, Australia 6 Faculty of Medicine, School of Women’s and Children’s Health, University of New South Wales, Sydney, NSW, Australia and Research Portfolio, University of Sydney, Sydney, NSW, Australia * These authors have contributed equally to this work Correspondence to: Jamie I. Fletcher, email: // Lesley J. Ashton, email: // Keywords : MYCN, MYC, SLC19A1, methotrexate, neuroblastoma Received : February 10, 2015 Accepted : March 10, 2015 Published : March 30, 2015 Abstract MYCN amplification occurs in 20% of neuroblastomas and is strongly related to poor clinical outcome. We have identified folate-mediated one-carbon metabolism as highly upregulated in neuroblastoma tumors with MYCN amplification and have validated this finding experimentally by showing that MYCN amplified neuroblastoma cell lines have a higher requirement for folate and are significantly more sensitive to the antifolate methotrexate than cell lines without MYCN amplification. We have demonstrated that methotrexate uptake in neuroblastoma cells is mediated principally by the reduced folate carrier (RFC; SLC19A1 ), that SLC19A1 and MYCN expression are highly correlated in both patient tumors and cell lines, and that SLC19A1 is a direct transcriptional target of N-Myc. Finally, we assessed the relationship between SLC19A1 expression and patient survival in two independent primary tumor cohorts and found that SLC19A1 expression was associated with increased risk of relapse or death, and that SLC19A1 expression retained prognostic significance independent of age, disease stage and MYCN amplification. This study adds upregulation of folate-mediated one-carbon metabolism to the known consequences of MYCN amplification, and suggests that this pathway might be targeted in poor outcome tumors with MYCN amplification and high SLC19A1 expression.
48. Expression Profiling of ABC Transporter Genes in Chronic Myeloid Leukemia (CML) and Responsiveness to Imatinib
- Author
-
Roberto Bernardoni, Samuele Gherardi, Giuseppe Saglio, Fabrizio Pane, Nunzio Iraci, Enrico Gottardi, Nicola Esposito, Daniela Cilloni, Gianantonio Rosti, Giovanni Perini, Simona Soverini, Francesca Palandri, Giovanni Martinelli, Fausto Castagnetti, Fabrizio Quarantelli, Emanuele Valli, Gabriele Gugliotta, Michele Baccarani, and Giuliano Della Valle
- Subjects
biology ,Immunology ,Imatinib ,ATP-binding cassette transporter ,Cell Biology ,Hematology ,ABCC4 ,Bioinformatics ,Biochemistry ,Gene expression profiling ,Imatinib mesylate ,ABCC3 ,Reference genes ,biology.protein ,Cancer research ,medicine ,Chromatin immunoprecipitation ,medicine.drug - Abstract
The deregulation of ATP-binding cassette (ABC) transporters responsible for the efflux of anticancer agents may involve mutations or single nucleotide polymorphisms (SNPs) or an increase in their expression level. Consequently, chemoresistance may develop. We have previously shown that the expression level and transcription of ABC drug transporters in CML cells is affected by c-Myc. Our results demonstrated that c-Myc is highly expressed in CD34+ cells from newly diagnosed chronic phase (CP)-CML patients, and that it can significantly upregulate the expression of several ABC genes, particularly, the ABCC1, ABCC4 and ABCG2, while it downregulates the expression of ABCC3. We have also demonstrated that c-Myc was a direct regulator and physically associated with the promoter of tested ABC genes, as assessed by Chromatin Immunoprecipitation in a cell line derived from a Ph+ CML patient. Taken together, our findings supported the model of a direct and coordinate regulation of a large set of ABC genes by the c-Myc transcription factor. Our study also supported prior findings that deregulation of specific set of ABC genes could be an important molecular mechanism altering imatinib transport. Based on these observations we have started to investigate the role of ABC transport genes expression in newly diagnosed CP-CML patients treated with imatinib. RNA extracted from white blood cells of 5 patients who achieved a stable major molecular response (MMR) by 12 months (responders) and 15 patients who didn’t show a partial cytogenetic response (CgR) by 6 months nor a complete CgR by 12 months (suboptimal responders according to European LeukemiaNet recommendations). All pts were enrolled on GIMEMA CML Working Party-sponsored clinical trials of imatinib. A panel of ABC genes including ABCB1, ABCB9, ABCC1, ABCC3, ABCC4, ABCE1 and ABCG2 was interrogated by Q-PCR for the level of expression. Results were normalized to the expression of three reference genes, i.e., GUSB, b-actin and GAPDH. Our results show that suboptimal responders display high expression levels of ABCG2 (p
49. A genome-scale shRNA screen identifies GSK3 as a critical regulator of p75NTR transcription in high risk neuroblastoma
- Author
-
PERINI, GIOVANNI, VALLI, EMANUELE, GHERARDI, SAMUELE, MILAZZO, GIORGIO, DI GIACOMO, SIMONE, Timothy R. Wagenaar, Michael R. Green, Giovanni Perini, Emanuele Valli, Samuele Gherardi, Simone Di Giacomo, Giorgio Milazzo, Timothy R. Wagenaar, and Michael R. Green
- Subjects
animal structures ,macromolecular substances ,sense organs - Abstract
Our findings support a model in which the GSK3 pathway can exerts a negative control on p75NTR transcription in high risk neuroblastoma and highlight the possibility to use GSK3 inhibitors as potential therapeutic drugs to treat neuroblastoma with low p75NTR.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.