160 results on '"Foiani, M."'
Search Results
2. The Rad53CHK1/CHK2-Spt21NPAT and Tel1ATM axes couple glucose tolerance to histone dosage and subtelomeric silencing
- Author
-
Bruhn, C, Ajazi, A, Ferrari, E, Lanz, M, Batrin, R, Choudhary, R, Walvekar, A, Laxman, S, Longhese, M, Fabre, E, Smolka, M, Foiani, M, Bruhn C., Ajazi A., Ferrari E., Lanz M. C., Batrin R., Choudhary R., Walvekar A., Laxman S., Longhese M. P., Fabre E., Smolka M. B., Foiani M., Bruhn, C, Ajazi, A, Ferrari, E, Lanz, M, Batrin, R, Choudhary, R, Walvekar, A, Laxman, S, Longhese, M, Fabre, E, Smolka, M, Foiani, M, Bruhn C., Ajazi A., Ferrari E., Lanz M. C., Batrin R., Choudhary R., Walvekar A., Laxman S., Longhese M. P., Fabre E., Smolka M. B., and Foiani M.
- Abstract
The DNA damage response (DDR) coordinates DNA metabolism with nuclear and non-nuclear processes. The DDR kinase Rad53CHK1/CHK2 controls histone degradation to assist DNA repair. However, Rad53 deficiency causes histone-dependent growth defects in the absence of DNA damage, pointing out unknown physiological functions of the Rad53-histone axis. Here we show that histone dosage control by Rad53 ensures metabolic homeostasis. Under physiological conditions, Rad53 regulates histone levels through inhibitory phosphorylation of the transcription factor Spt21NPAT on Ser276. Rad53-Spt21 mutants display severe glucose dependence, caused by excess histones through two separable mechanisms: dampening of acetyl-coenzyme A-dependent carbon metabolism through histone hyper-acetylation, and Sirtuin-mediated silencing of starvation-induced subtelomeric domains. We further demonstrate that repression of subtelomere silencing by physiological Tel1ATM and Rpd3HDAC activities coveys tolerance to glucose restriction. Our findings identify DDR mutations, histone imbalances and aberrant subtelomeric chromatin as interconnected causes of glucose dependence, implying that DDR kinases coordinate metabolism and epigenetic changes.
- Published
- 2020
3. Muscle and not neuronal biomarkers correlate with severity in spinal and bulbar muscular atrophy
- Author
-
Lombardi, V, Querin, G, Ziff, O, Zampedri, L, Martinelli, I, Heller, C, Foiani, M, Bertolin, C, Lu, C, Malik, B, Allen, K, Rinaldi, C, Zetterberg, H, Heslegrave, A, Greensmith, L, Hanna, M, Soraru, G, Malaspina, A, and Fratta, P
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Bulbo-Spinal Atrophy, X-Linked ,Severity of Illness Index ,Article ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Neurofilament Proteins ,medicine ,Animals ,Humans ,Longitudinal Studies ,Prospective Studies ,030212 general & internal medicine ,Creatine Kinase ,Aged ,Creatinine ,business.industry ,Amyotrophic Lateral Sclerosis ,Correction ,Middle Aged ,medicine.disease ,Disease Models, Animal ,Spinal and bulbar muscular atrophy ,nervous system ,chemistry ,Female ,Neurology (clinical) ,business ,Biomarkers ,030217 neurology & neurosurgery - Abstract
Objective To determine whether blood biomarkers of neuronal damage (neurofilament light chain [NfL]), muscle damage (creatine kinase [CK]), and muscle mass (creatinine) are altered in spinal and bulbar muscular atrophy (SBMA) and can be used as biomarkers for disease severity. Methods In this multicenter longitudinal prospective study, plasma and serum were collected from 2 cohorts of patients with SBMA in London, United Kingdom (n = 50), and Padova, Italy (n = 43), along with disease (amyotrophic lateral sclerosis [ALS]) and healthy controls, and levels of plasma and serum NfL, CK, and creatinine were measured. Disease severity was assessed by the SBMA Functional Rating Scale and the Adult Myopathy Assessment Tool at baseline and 12 and 24 months. Results Blood NfL concentrations were increased in ALS samples, but were unchanged in both SBMA cohorts, were stable after 12 and 24 months, and were not correlated with clinical severity. Normal NfL levels were also found in a well-established mouse model of SBMA. Conversely, CK concentrations were significantly raised in SBMA compared with ALS samples, and were not correlated to the clinical measures. Creatinine concentrations were significantly reduced in SBMA, and strongly and significantly correlated with disease severity. Conclusions While muscle damage and muscle mass biomarkers are abnormal in SBMA, axonal damage markers are unchanged, highlighting the relevant primary role of skeletal muscle in disease pathogenesis. Creatinine, but not CK, correlated with disease severity, confirming its role as a valuable biomarker in SBMA.
- Published
- 2020
4. 248P Targeting triple-negative breast cancer metabolism with neoadjuvant chemotherapy plus fasting-mimicking diet plus/minus metformin: The BREAKFAST trial
- Author
-
Ligorio, F., Fucà, G., Vingiani, A., Iannelli, F., Lobefaro, R., Ferraris, C., Belfiore, A., Scaperrotta, G., Depretto, C., Martinetti, A., Corsetto, P., Bianchi, G.V., Capri, G., Folli, S., Minucci, S., Foiani, M., Pagani, M., Pruneri, G., De Braud, F.G.M., and Vernieri, C.
- Published
- 2023
- Full Text
- View/download PDF
5. Topological stress and genome integrity maintenance: P21-7
- Author
-
Capra, T., Jossen, R., Colosio, A., Frattini, C., Aguilera, A., Shirahige, K., Foiani, M., and Bermejo, R.
- Published
- 2012
6. White matter hyperintensities in progranulin-associated frontotemporal dementia: A longitudinal GENFI study
- Author
-
Sudre, C.H. (Carole H.), Bocchetta, M. (Martina), Heller, C. (Carolin), Convery, R. (Rhian), Neason, M. (Mollie), Moore, K.M. (Katrina M.), Cash, D.M. (David M.), Thomas, D.L. (David L), Woollacott, I.O.C. (Ione O.C.), Foiani, M. (Martha), Heslegrave, A. (Amanda), Shafei, R. (Rachelle), Greaves, C. (Caroline), Swieten, J.C. (John) van, Moreno, F. (Fermin), Sánchez-Valle, R. (Raquel), Borroni, B. (Barbara), Laforce, R. (Robert), Masellis, M. (Mario), Tartaglia, M.C. (Maria Carmela), Graff, C. (Caroline), Galimberti, D. (Daniela), Rowe, J.B. (James), Finger, E. (Elizabeth), Synofzik, M. (Matthis), Vandenberghe, R. (Rik), De Mendonça, A. (Alexandre), Tagliavini, F. (Fabrizio), Santana, I. (Isabel), Ducharme, S. (Simon), Butler, C. (Chris), Gerhard, A. (Alex), Levin, J. (Johannes), Danek, A. (Adrian), Frisoni, G.B. (Giovanni B.), Sorbi, S. (Sandro), Otto, M. (Markus), Zetterberg, H. (Henrik), Ourselin, S. (Sebastien), Cardoso, M.J. (M. Jorge), Rohrer, J.D. (Jonathan), Sudre, C.H. (Carole H.), Bocchetta, M. (Martina), Heller, C. (Carolin), Convery, R. (Rhian), Neason, M. (Mollie), Moore, K.M. (Katrina M.), Cash, D.M. (David M.), Thomas, D.L. (David L), Woollacott, I.O.C. (Ione O.C.), Foiani, M. (Martha), Heslegrave, A. (Amanda), Shafei, R. (Rachelle), Greaves, C. (Caroline), Swieten, J.C. (John) van, Moreno, F. (Fermin), Sánchez-Valle, R. (Raquel), Borroni, B. (Barbara), Laforce, R. (Robert), Masellis, M. (Mario), Tartaglia, M.C. (Maria Carmela), Graff, C. (Caroline), Galimberti, D. (Daniela), Rowe, J.B. (James), Finger, E. (Elizabeth), Synofzik, M. (Matthis), Vandenberghe, R. (Rik), De Mendonça, A. (Alexandre), Tagliavini, F. (Fabrizio), Santana, I. (Isabel), Ducharme, S. (Simon), Butler, C. (Chris), Gerhard, A. (Alex), Levin, J. (Johannes), Danek, A. (Adrian), Frisoni, G.B. (Giovanni B.), Sorbi, S. (Sandro), Otto, M. (Markus), Zetterberg, H. (Henrik), Ourselin, S. (Sebastien), Cardoso, M.J. (M. Jorge), and Rohrer, J.D. (Jonathan)
- Abstract
Frontotemporal dementia (FTD) is a heterogeneous group of neurodegenerative disorders with both sporadic and genetic forms. Mutations in the progranulin gene (GRN) are a common cause of genetic FTD, causing either a behavioural presentation or, less commonly, language impairment. Presence on T2-weighted images of white matter hyperintensities (WMH) has been previously shown to be more commonly associated with GRN mutations rather than other forms of FTD. The aim of the current study was to investigate the longitudinal change in WMH and the associations of WMH burden with grey matter (GM) loss, markers of neurodegeneration and cognitive function in GRN mutation carriers. 336 participants in the Genetic FTD Initiative (GENFI) study were included in the analysis: 101 presymptomatic and 32 symptomatic GRN mutation carriers, as well as 203 mutation-negative controls. 39 presymptomatic and 12 symptomatic carriers, and 73 controls also had longitudinal data available. Participants underwent MR imaging acquisition including isotropic 1 mm T1-weighted and T2-weighted sequences. WMH were automatically segmented and locally subdivided to enable a more detailed representation of the pathology distribution. Log-transformed WMH volumes were investigated in terms of their global and regional associations with imaging measures (grey matter volumes), biomarker concentrations (plasma neurofilament light chain, NfL, and glial fibrillary acidic protein, GFAP), genetic status (TMEM106B risk genotype) and cognition (tests of executive function). Analyses revealed that WMH load was higher in both symptomatic and presymptomatic groups compared with controls and this load increased over time. In particular, lesions were seen periventricularly in frontal and occipital lobes, progressing to medial layers over time. However, there was variability in the WMH load across GRN mutation carriers – in the symptomatic group 25.0% had none/mild load, 37.5% had medium and 37.5% had a severe load – a di
- Published
- 2019
- Full Text
- View/download PDF
7. PDXK mutations cause polyneuropathy responsive to pyridoxal 5'-phosphate supplementation
- Author
-
Chelban, V., Wilson, M. P., Warman Chardon, J., Vandrovcova, J., Zanetti, M. N., Zamba-Papanicolaou, E., Efthymiou, S., Pope, S., Conte, M. R., Abis, G., Liu, Y. -T., Tribollet, E., Haridy, N. A., Botia, J. A., Ryten, M., Nicolaou, P., Minaidou, A., Christodoulou, K., Kernohan, K. D., Eaton, A., Osmond, M., Ito, Y., Bourque, P., Jepson, J. E. C., Bello, O., Bremner, F., Cordivari, C., Reilly, M. M., Foiani, M., Heslegrave, A., Zetterberg, H., Heales, S. J. R., Wood, N. W., Rothman, J. E., Boycott, K. M., Mills, P. B., Clayton, P. T., Houlden, H., Kriouile, Y., Khorassani, M. E., Aguennouz, M., Groppa, S., Marinova Karashova, B., Van Maldergem, L., Nachbauer, W., Boesch, S., Arning, L., Timmann, D., Cormand, B., Perez-Duenas, B., Di Rosa, G., Goraya, J. S., Sultan, T., Mine, J., Avdjieva, D., Kathom, H., Tincheva, R., Banu, S., Pineda-Marfa, M., Veggiotti, P., Ferrari, M. D., van den Maagdenberg, A. M. J. M., Verrotti, A., Marseglia, G., Savasta, S., Garcia-Silva, M., Ruiz, A. M., Garavaglia, B., Borgione, E., Portaro, S., Sanchez, B. M., Boles, R., Papacostas, S., Vikelis, M., Giunti, P., Salpietro, V., Oconnor, E., Kullmann, D., Kaiyrzhanov, R., Sullivan, R., Khan, A. M., Yau, W. Y., Hostettler, I., Papanicolaou, E. Z., Dardiotis, E., Maqbool, S., Ibrahim, S., Kirmani, S., Rana, N. N., Atawneh, O., Lim, S. -Y., Shaikh, F., Koutsis, G., Breza, M., Mangano, S., Scuderi, C., Morello, G., Stojkovic, T., Torti, E., Zollo, M., Heimer, G., Dauvilliers, Y. A., Striano, P., Al-Khawaja, I., Al-Mutairi, F., Alkuraya, F. S., Sherifa, H., Rizig, M., Okubadejo, N. U., Ojo, O. O., Oshinaike, O. O., Wahab, K., Bello, A. H., Abubakar, S., Obiabo, Y., Nwazor, E., Ekenze, O., Williams, U., Iyagba, A., Taiwo, L., Komolafe, M., Oguntunde, O., Pchelina, S., Senkevich, K., Shashkin, C., Zharkynbekova, N., Koneyev, K., Manizha, G., Isrofilov, M., Guliyeva, U., Salayev, K., Khachatryan, S., Rossi, S., Silvestri, Gabriella, Bourinaris, T., Xiromerisiou, G., Fidani, L., Spanaki, C., Tucci, A., Silvestri G. (ORCID:0000-0002-1950-1468), Chelban, V., Wilson, M. P., Warman Chardon, J., Vandrovcova, J., Zanetti, M. N., Zamba-Papanicolaou, E., Efthymiou, S., Pope, S., Conte, M. R., Abis, G., Liu, Y. -T., Tribollet, E., Haridy, N. A., Botia, J. A., Ryten, M., Nicolaou, P., Minaidou, A., Christodoulou, K., Kernohan, K. D., Eaton, A., Osmond, M., Ito, Y., Bourque, P., Jepson, J. E. C., Bello, O., Bremner, F., Cordivari, C., Reilly, M. M., Foiani, M., Heslegrave, A., Zetterberg, H., Heales, S. J. R., Wood, N. W., Rothman, J. E., Boycott, K. M., Mills, P. B., Clayton, P. T., Houlden, H., Kriouile, Y., Khorassani, M. E., Aguennouz, M., Groppa, S., Marinova Karashova, B., Van Maldergem, L., Nachbauer, W., Boesch, S., Arning, L., Timmann, D., Cormand, B., Perez-Duenas, B., Di Rosa, G., Goraya, J. S., Sultan, T., Mine, J., Avdjieva, D., Kathom, H., Tincheva, R., Banu, S., Pineda-Marfa, M., Veggiotti, P., Ferrari, M. D., van den Maagdenberg, A. M. J. M., Verrotti, A., Marseglia, G., Savasta, S., Garcia-Silva, M., Ruiz, A. M., Garavaglia, B., Borgione, E., Portaro, S., Sanchez, B. M., Boles, R., Papacostas, S., Vikelis, M., Giunti, P., Salpietro, V., Oconnor, E., Kullmann, D., Kaiyrzhanov, R., Sullivan, R., Khan, A. M., Yau, W. Y., Hostettler, I., Papanicolaou, E. Z., Dardiotis, E., Maqbool, S., Ibrahim, S., Kirmani, S., Rana, N. N., Atawneh, O., Lim, S. -Y., Shaikh, F., Koutsis, G., Breza, M., Mangano, S., Scuderi, C., Morello, G., Stojkovic, T., Torti, E., Zollo, M., Heimer, G., Dauvilliers, Y. A., Striano, P., Al-Khawaja, I., Al-Mutairi, F., Alkuraya, F. S., Sherifa, H., Rizig, M., Okubadejo, N. U., Ojo, O. O., Oshinaike, O. O., Wahab, K., Bello, A. H., Abubakar, S., Obiabo, Y., Nwazor, E., Ekenze, O., Williams, U., Iyagba, A., Taiwo, L., Komolafe, M., Oguntunde, O., Pchelina, S., Senkevich, K., Shashkin, C., Zharkynbekova, N., Koneyev, K., Manizha, G., Isrofilov, M., Guliyeva, U., Salayev, K., Khachatryan, S., Rossi, S., Silvestri, Gabriella, Bourinaris, T., Xiromerisiou, G., Fidani, L., Spanaki, C., Tucci, A., and Silvestri G. (ORCID:0000-0002-1950-1468)
- Abstract
Objective: To identify disease-causing variants in autosomal recessive axonal polyneuropathy with optic atrophy and provide targeted replacement therapy. Methods: We performed genome-wide sequencing, homozygosity mapping, and segregation analysis for novel disease-causing gene discovery. We used circular dichroism to show secondary structure changes and isothermal titration calorimetry to investigate the impact of variants on adenosine triphosphate (ATP) binding. Pathogenicity was further supported by enzymatic assays and mass spectroscopy on recombinant protein, patient-derived fibroblasts, plasma, and erythrocytes. Response to supplementation was measured with clinical validated rating scales, electrophysiology, and biochemical quantification. Results: We identified biallelic mutations in PDXK in 5 individuals from 2 unrelated families with primary axonal polyneuropathy and optic atrophy. The natural history of this disorder suggests that untreated, affected individuals become wheelchair-bound and blind. We identified conformational rearrangement in the mutant enzyme around the ATP-binding pocket. Low PDXK ATP binding resulted in decreased erythrocyte PDXK activity and low pyridoxal 5′-phosphate (PLP) concentrations. We rescued the clinical and biochemical profile with PLP supplementation in 1 family, improvement in power, pain, and fatigue contributing to patients regaining their ability to walk independently during the first year of PLP normalization. Interpretation: We show that mutations in PDXK cause autosomal recessive axonal peripheral polyneuropathy leading to disease via reduced PDXK enzymatic activity and low PLP. We show that the biochemical profile can be rescued with PLP supplementation associated with clinical improvement. As B6 is a cofactor in diverse essential biological pathways, our findings may have direct implications for neuropathies of unknown etiology characterized by reduced PLP levels. ANN NEUROL 2019;86:225–240.
- Published
- 2019
8. PP2A Controls Genome Integrity by Integrating Nutrient-Sensing and Metabolic Pathways with the DNA Damage Response
- Author
-
Ferrari, E, Bruhn, C, Peretti, M, Cassani, C, Carotenuto, W, Elgendy, M, Shubassi, G, Lucca, C, Bermejo, R, Varasi, M, Minucci, S, Longhese, M, Foiani, M, Carotenuto, WV, Longhese, MP, Ferrari, E, Bruhn, C, Peretti, M, Cassani, C, Carotenuto, W, Elgendy, M, Shubassi, G, Lucca, C, Bermejo, R, Varasi, M, Minucci, S, Longhese, M, Foiani, M, Carotenuto, WV, and Longhese, MP
- Abstract
Mec1ATR mediates the DNA damage response (DDR), integrating chromosomal signals and mechanical stimuli. We show that the PP2A phosphatases, ceramide-activated enzymes, couple cell metabolism with the DDR. Using genomic screens, metabolic analysis, and genetic and pharmacological studies, we found that PP2A attenuates the DDR and that three metabolic circuits influence the DDR by modulating PP2A activity. Irc21, a putative cytochrome b5 reductase that promotes the condensation reaction generating dihydroceramides (DHCs), and Ppm1, a PP2A methyltransferase, counteract the DDR by activating PP2A; conversely, the nutrient-sensing TORC1-Tap42 axis sustains DDR activation by inhibiting PP2A. Loss-of-function mutations in IRC21, PPM1, and PP2A and hyperactive tap42 alleles rescue mec1 mutants. Ceramides synergize with rapamycin, a TORC1 inhibitor, in counteracting the DDR. Hence, PP2A integrates nutrient-sensing and metabolic pathways to attenuate the Mec1ATR response. Our observations imply that metabolic changes affect genome integrity and may help with exploiting therapeutic options and repositioning known drugs.
- Published
- 2017
9. Chromatin replication and histone dynamics
- Author
-
Masai, H., Foiani, M., Alabert, Constance, Jasencakova, Zuzana, Groth, Anja, Masai, H., Foiani, M., Alabert, Constance, Jasencakova, Zuzana, and Groth, Anja
- Abstract
Inheritance of the DNA sequence and its proper organization into chromatin is fundamental for genome stability and function. Therefore, how specific chromatin structures are restored on newly synthesized DNA and transmitted through cell division remains a central question to understand cell fate choices and self-renewal. Propagation of genetic information and chromatin-based information in cycling cells entails genome-wide disruption and restoration of chromatin, coupled with faithful replication of DNA. In this chapter, we describe how cells duplicate the genome while maintaining its proper organization into chromatin. We reveal how specialized replication-coupled mechanisms rapidly assemble newly synthesized DNA into nucleosomes, while the complete restoration of chromatin organization including histone marks is a continuous process taking place throughout the cell cycle. Because failure to reassemble nucleosomes at replication forks blocks DNA replication progression in higher eukaryotes and leads to genomic instability, we further underline the importance of the mechanistic link between DNA replication and chromatin duplication.
- Published
- 2017
10. Rad51-dependent DNA structures accumulate at damaged replication forks in sgs1 mutants defective in the yeast ortholog of BLM RecQ helicase
- Author
-
Liberi, G, Maffioletti, G, Lucca, C, Chiolo, I, Baryshnikova, A, Cotta-Ramusino, C, Lopes, Massimo; https://orcid.org/0000-0003-3847-8133, Pellicioli, A, Harber, J E, Foiani, M, Liberi, G, Maffioletti, G, Lucca, C, Chiolo, I, Baryshnikova, A, Cotta-Ramusino, C, Lopes, Massimo; https://orcid.org/0000-0003-3847-8133, Pellicioli, A, Harber, J E, and Foiani, M
- Abstract
S-phase cells overcome chromosome lesions through replication-coupled recombination processes that seem to be assisted by recombination-dependent DNA structures and/or replication-related sister chromatid junctions. RecQ helicases, including yeast Sgs1 and human BLM, have been implicated in both replication and recombination and protect genome integrity by preventing unscheduled mitotic recombination events. We have studied the RecQ helicase-mediated mechanisms controlling genome stability by analyzing replication forks encountering a damaged template in sgs1 cells. We show that, in sgs1 mutants, recombination-dependent cruciform structures accumulate at damaged forks. Their accumulation requires Rad51 protein, is counteracted by Srs2 DNA helicase, and does not prevent fork movement. Sgs1, but not Srs2, promotes resolution of these recombination intermediates. A functional Rad53 checkpoint kinase that is known to protect the integrity of the sister chromatid junctions is required for the accumulation of recombination intermediates in sgs1 mutants. Finally, top3 and top3 sgs1 mutants accumulate the same structures as sgs1 cells. We suggest that, in sgs1 cells, the unscheduled accumulation of Rad51-dependent cruciform structures at damaged forks result from defective maturation of recombination-dependent intermediates that originate from the replication-related sister chromatid junctions. Our findings might contribute to explaining some of the recombination defects of BLM cells.
- Published
- 2005
11. Leaping forks at inverted repeats
- Author
-
Branzei, D., primary and Foiani, M., additional
- Published
- 2010
- Full Text
- View/download PDF
12. Dissection of RNA Binding Domains from Yeast NSR1 Protein
- Author
-
Gamberi, C., Romanelli, Maria, Contreas, G., Foiani, M., and Morandi, Carlo
- Published
- 1993
13. La Proteina nucleolare NSR1 di lievito: domini di 'binding' per l'RNA e confronto con l'hnRNP di tipo A1 umana
- Author
-
Romanelli, Maria, Gamberi, C., Contreas, G., Foiani, M., and Morandi, Carlo
- Published
- 1993
14. A dominant-negative MEC3 mutant uncovers new functions for the Rad17 complex and Tel1
- Author
-
Giannattasio, M., primary, Sommariva, E., additional, Vercillo, R., additional, Lippi-Boncambi, F., additional, Liberi, G., additional, Foiani, M., additional, Plevani, P., additional, and Muzi-Falconi, M., additional
- Published
- 2002
- Full Text
- View/download PDF
15. Spk1/Rad53 is regulated by Mec1-dependent protein phosphorylation in DNA replication and damage checkpoint pathways.
- Author
-
Sun, Z, primary, Fay, D S, additional, Marini, F, additional, Foiani, M, additional, and Stern, D F, additional
- Published
- 1996
- Full Text
- View/download PDF
16. Cell cycle-dependent phosphorylation and dephosphorylation of the yeast DNA polymerase alpha-primase B subunit
- Author
-
Foiani, M, primary, Liberi, G, additional, Lucchini, G, additional, and Plevani, P, additional
- Published
- 1995
- Full Text
- View/download PDF
17. The B subunit of the DNA polymerase alpha-primase complex in Saccharomyces cerevisiae executes an essential function at the initial stage of DNA replication
- Author
-
Foiani, M, primary, Marini, F, additional, Gamba, D, additional, Lucchini, G, additional, and Plevani, P, additional
- Published
- 1994
- Full Text
- View/download PDF
18. De novo synthesis of budding yeast DNA polymerase alpha and POL1 transcription at the G1/S boundary are not required for entrance into S phase.
- Author
-
Falconi, M M, primary, Piseri, A, additional, Ferrari, M, additional, Lucchini, G, additional, Plevani, P, additional, and Foiani, M, additional
- Published
- 1993
- Full Text
- View/download PDF
19. Guanine nucleotide exchange factor for eukaryotic translation initiation factor 2 in Saccharomyces cerevisiae: interactions between the essential subunits GCD2, GCD6, and GCD7 and the regulatory subunit GCN3.
- Author
-
Bushman, J L, primary, Foiani, M, additional, Cigan, A M, additional, Paddon, C J, additional, and Hinnebusch, A G, additional
- Published
- 1993
- Full Text
- View/download PDF
20. The isolated 48,000-dalton subunit of yeast DNA primase is sufficient for RNA primer synthesis.
- Author
-
Santocanale, C., primary, Foiani, M., additional, Lucchini, G., additional, and Plevani, P., additional
- Published
- 1993
- Full Text
- View/download PDF
21. GCD2, a translational repressor of the GCN4 gene, has a general function in the initiation of protein synthesis in Saccharomyces cerevisiae
- Author
-
Foiani, M, primary, Cigan, A M, additional, Paddon, C J, additional, Harashima, S, additional, and Hinnebusch, A G, additional
- Published
- 1991
- Full Text
- View/download PDF
22. Complex formation by positive and negative translational regulators of GCN4.
- Author
-
Cigan, A M, primary, Foiani, M, additional, Hannig, E M, additional, and Hinnebusch, A G, additional
- Published
- 1991
- Full Text
- View/download PDF
23. Yeast DNA polymerase‐‐DNA primase complex; cloning of PRI 1, a single essential gene related to DNA primase activity.
- Author
-
Lucchini, G., Francesconi, S., Foiani, M., Badaracco, G., and Plevani, P.
- Abstract
The immunopurified yeast DNA polymerase‐‐DNA primase complex is constituted by DNA polymerase I polypeptides and by three other protein species, called p74, p58 and p48, which we show to be immunologically unrelated. The gene encoding the p48 polypeptide has been identified by immunological screening of a lambda gt11 yeast genomic DNA library. Antiserum specific for p48 inhibits DNA primase, and immunoreactive, inhibitory antibodies are affinity‐purified by the clone‐encoded protein, thus relating the p48 polypeptide to DNA primase activity. The entire gene has been cloned, and the 1.45‐kb p48 mRNA is overproduced in cells containing the gene in high copy number. Gene disruption and Southern hybridization experiments demonstrate that the p48 protein is encoded by a single gene and it performs an essential function.
- Published
- 1987
- Full Text
- View/download PDF
24. A single essential gene, PRI2, encodes the large subunit of DNA primase in Saccharomyces cerevisiae
- Author
-
Foiani, M, Santocanale, C, Plevani, P, and Lucchini, G
- Abstract
DNA primase activity of the yeast DNA polymerase-primase complex is related to two polypeptides, p58 and p48. The reciprocal role of these protein species has not yet been clarified, although both participate in formation of the active center of the enzyme. The gene encoding the p58 subunit has been cloned by screening of a lambda gt11 yeast genomic DNA library, using specific anti-p58 antiserum. Antibodies that inhibited DNA primase activity could be purified by lysates of Escherichia coli cells infected with a recombinant bacteriophage containing the entire gene, which we designate PR12. The gene was found to be transcribed in a 1.7-kilobase mRNA whose level appeared to fluctuate during the mitotic cell cycle. Nucleotide sequence determination indicated that PR12 encodes a 528-amino-acid polypeptide with a calculated molecular weight of 62,262. The gene is unique in the haploid yeast genome, and its product is essential for cell viability, as has been shown for other components of the yeast DNA polymerase-primase complex.
- Published
- 1989
- Full Text
- View/download PDF
25. Phosphorylation of the DNA polymerase alpha-primase B subunit is dependent on its association with the p180 polypeptide.
- Author
-
Ferrari, M, Lucchini, G, Plevani, P, and Foiani, M
- Abstract
The B subunit of the DNA polymerase (pol) alpha-primase complex executes an essential role at the initial stage of DNA replication in Saccharomyces cerevisiae and is phosphorylated in a cell cycle-dependent manner. In this report, we show that the four subunits of the yeast DNA polymerase alpha-primase complex are assembled throughout the cell cycle, and physical association between newly synthesized pol alpha (p180) and unphosphorylated B subunit (p86) occurs very rapidly. Therefore, B subunit phosphorylation does not appear to modulate p180.p86 interaction. Conversely, by depletion experiments and by using a yeast mutant strain, which produces a low and constitutive level of the p180 polypeptide, we found that formation of the p180.p86 subcomplex is required for B subunit phosphorylation.
- Published
- 1996
26. Polypeptide structure of DNA primase from a yeast DNA polymerase-primase complex.
- Author
-
Plevani, P, Foiani, M, Valsasnini, P, Badaracco, G, Cheriathundam, E, and Chang, L M
- Abstract
An immunoaffinity chromatographic procedure was developed to purify DNA polymerase-DNA primase complex from crude soluble extracts of yeast cells. The immunoabsorbent column is made of mouse monoclonal antibody to yeast DNA polymerase I covalently linked to Protein A-Sepharose. Purification of the complex involves binding of the complex to the immunoabsorbent column and elution with concentrated MgCl2 solutions. After rebinding to the monoclonal antibody column free primase activity is selectively eluted with a lower concentration of MgCl2. Polyacrylamide gel electrophoresis in the presence of sodium dodecyl sulfate showed the presence of five major peptides, p180, p140, p74, p58, and p48 in the immunoaffinity-purified DNA polymerase-DNA primase complex. Free primase and free polymerase fractions obtained by fractionation on the immunoabsorbent column were analyzed on activity gels and immunoblots. These analyses showed that p180 and p140 are DNA polymerase peptides. Two polypeptides of 58 and 48 kDa co-fractionated with the free yeast DNA primase. From sucrose gradient analysis we estimate a molecular weight of 110 kDa for the native DNA primase.
- Published
- 1985
- Full Text
- View/download PDF
27. Affinity Labeling of the Active Center and Ribonucleoside Triphosphate Binding Site of Yeast DNA Primase
- Author
-
Foiani, M, Lindner, A J, Hartmann, G R, Lucchini, G, and Plevani, P
- Abstract
A highly selective affinity labeling procedure has been applied to map the active center of DNA primase from the yeast Saccharomyces cerevisiae. Enzyme molecules that have been modified by covalent attachment of benzaldehyde derivatives of adenine nucleotides are autocatalytically labeled by incubation with a radioactive ribonucleoside triphosphate. The affinity labeling of primase requires a template DNA, is not affected by DNase and RNase treatments, but is sensitive to proteinase K. Both the p58 and p48 subunits of yeast DNA primase appear to participate in the formation of the catalytic site of the enzyme, although UV-photocross-linking with [α-32P]ATP locates the ribonucleoside triphosphate binding site exclusively on the p48 polypeptide. The fixation of the radioactive product has been carried out also after the enzymatic reaction. Under this condition the RNA primers synthesized by the DNA polymerase-primase complex under uncoupled DNA synthesis conditions are linked to both DNA primase and DNA polymerase. When DNA synthesis is allowed to proceed first, the labeled RNA chains are fixed exclusively to the DNA polymerase polypeptide. These results, in accord with previous data, have been used to propose a model illustrating the interactions and the putative roles of the polypeptides of the DNA polymerase-primase complex.
- Published
- 1989
- Full Text
- View/download PDF
28. Identification of an effective chemotherapy and DNA damage response inhibitor combination for diffuse large b cell lymphoma
- Author
-
A. Chan, A. Anbuselvan, S.S. Upadhyayula, S. Jemimah, P. Jaynes, M.M. Hoppe, J.D. Wardyn, J. Goh, G. Bertolazzi, M. Foiani, M.J. O’Connor, E.K. Chow, C. Tripodo, A.D. Jeyasekharan, Chan, A., Anbuselvan, A., Upadhyayula, S.S., Jemimah, S., Jaynes, P., Hoppe, M.M., Wardyn, J.D., Goh, J., Bertolazzi, G., Foiani, M., O’Connor, M.J., Chow, E.K., Tripodo, C., and Jeyasekharan, A.D.
- Subjects
Cancer Research ,Oncology ,DLBCL, DNA damage, chemotherapy ,Settore MED/42 - Igiene Generale E Applicata ,Settore MED/01 - Statistica Medica - Abstract
Chemotherapy forms the backbone of treatment for Diffuse Large B Cell Lymphoma (DLBCL); however
- Published
- 2022
29. The Rad53CHK1/CHK2-Spt21NPAT and Tel1ATM axes couple glucose tolerance to histone dosage and subtelomeric silencing
- Author
-
Bruhn, Christopher, Ajazi, Arta, Ferrari, Elisa, Lanz, Michael Charles, Lanz, Michael, Batrin, Renaud, Choudhary, Ramveer, Walvekar, Adhish, Laxman, Sunil, Longhese, Maria Pia, Fabre, Emmanuelle, Bustamente Smolka, Marcus, Foiani, Marco, IFOM, Istituto FIRC di Oncologia Molecolare (IFOM), Cornell University [New York], Génomes, biologie cellulaire et thérapeutiques (GenCellDi (UMR_S_944)), Collège de France (CdF (institution))-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), Institute for Stem Cell Science and Regenerative Medicine [Bangalore, Inde] (inStem), Università degli Studi di Milano-Bicocca [Milano] (UNIMIB), Università degli Studi di Milano [Milano] (UNIMI), C.B. was supported by fellowships from Associazione Italiana per la Ricerca sul Cancro (AIRC) Fellowship i-Care (Marie Curie co-funded by the European Union, ID 16173) and the European Commission (EC-FP7-SIPOD, ID PCOFUND-GA-2012-600399). This work was supported by grants from Fondazione AIRC under IG 2015 (M.F., ID 16770), IG 2018 (M.F., ID 21416), and IG 2017 (M.P.L., ID 19783), by the Ministero dell'Istruzione/Ministero dell'Università e della Ricerca (M.F., MIUR-PRIN-15-FOIANI) and by Progetti di Ricerca di Interesse Nazionale (PRIN) 2015 (M.P.L.). E. Fabre acknowledges Labex 'Who am I?' (ANR-11-LABX-0071, Idex ANR-11-IDEX-0005-02) and Cancéropôle Ile de France (ORFOCRISE PME-2015)., ANR-11-IDEX-0005,USPC,Université Sorbonne Paris Cité(2011), Bruhn, C, Ajazi, A, Ferrari, E, Lanz, M, Batrin, R, Choudhary, R, Walvekar, A, Laxman, S, Longhese, M, Fabre, E, Smolka, M, Foiani, M, Génomes, biologie cellulaire et thérapeutiques (GenCellDi (U944 / UMR7212)), Collège de France (CdF (institution))-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), Università degli Studi di Milano-Bicocca = University of Milano-Bicocca (UNIMIB), Università degli Studi di Milano = University of Milan (UNIMI), Bodescot, Myriam, and Université Sorbonne Paris Cité - - USPC2011 - ANR-11-IDEX-0005 - IDEX - VALID
- Subjects
0301 basic medicine ,DNA Repair ,DNA damage ,DNA repair ,[SDV]Life Sciences [q-bio] ,Science ,General Physics and Astronomy ,Saccharomyces cerevisiae ,[SDV.BC.BC]Life Sciences [q-bio]/Cellular Biology/Subcellular Processes [q-bio.SC] ,02 engineering and technology ,Protein-Serine-Threonine Kinase ,General Biochemistry, Genetics and Molecular Biology ,Ataxia Telangiectasia Mutated Protein ,03 medical and health sciences ,Histone Deacetylase ,Cell Cycle Protein ,[SDV.BC.BC] Life Sciences [q-bio]/Cellular Biology/Subcellular Processes [q-bio.SC] ,Serine ,Gene Silencing ,Epigenetics ,Phosphorylation ,lcsh:Science ,Transcription factor ,ComputingMilieux_MISCELLANEOUS ,Multidisciplinary ,biology ,Chemistry ,Acetylation ,General Chemistry ,Telomere ,021001 nanoscience & nanotechnology ,Subtelomere ,Chromatin ,Cell biology ,Checkpoint Kinase 2 ,Histone ,Glucose ,030104 developmental biology ,Intracellular Signaling Peptides and Protein ,Mutation ,biology.protein ,lcsh:Q ,0210 nano-technology ,Saccharomyces cerevisiae Protein ,DNA Damage ,Transcription Factors - Abstract
The DNA damage response (DDR) coordinates DNA metabolism with nuclear and non-nuclear processes. The DDR kinase Rad53CHK1/CHK2 controls histone degradation to assist DNA repair. However, Rad53 deficiency causes histone-dependent growth defects in the absence of DNA damage, pointing out unknown physiological functions of the Rad53-histone axis. Here we show that histone dosage control by Rad53 ensures metabolic homeostasis. Under physiological conditions, Rad53 regulates histone levels through inhibitory phosphorylation of the transcription factor Spt21NPAT on Ser276. Rad53-Spt21 mutants display severe glucose dependence, caused by excess histones through two separable mechanisms: dampening of acetyl-coenzyme A-dependent carbon metabolism through histone hyper-acetylation, and Sirtuin-mediated silencing of starvation-induced subtelomeric domains. We further demonstrate that repression of subtelomere silencing by physiological Tel1ATM and Rpd3HDAC activities coveys tolerance to glucose restriction. Our findings identify DDR mutations, histone imbalances and aberrant subtelomeric chromatin as interconnected causes of glucose dependence, implying that DDR kinases coordinate metabolism and epigenetic changes.
- Published
- 2020
- Full Text
- View/download PDF
30. ATR is essential for preservation of cell mechanics and nuclear integrity during interstitial migration
- Author
-
Kidiyoor, Gururaj Rao, Li, Qingsen, Bastianello, Giulia, Bruhn, Christopher, Giovannetti, Irene, Mohamood, Adhil, Beznoussenko, Galina V., Mironov, Alexandre, Raab, Matthew, Piel, Matthieu, Restuccia, Umberto, Matafora, Vittoria, Bachi, Angela, Barozzi, Sara, Parazzoli, Dario, Frittoli, Emanuela, Palamidessi, Andrea, Panciera, Tito, Piccolo, Stefano, Scita, Giorgio, Maiuri, Paolo, Havas, Kristina M., Zhou, Zhong-Wei, Kumar, Amit, Bartek, Jiri, Wang, Zhao-Qi, Foiani, Marco, Institut Curie [Paris], Kidiyoor, G R, Li, Q, Bastianello, G, Bruhn, C, Giovannetti, I, Mohamood, A, Beznoussenko, G V, Mironov, A, Raab, M, Piel, M, Restuccia, U, Matafora, V, Bachi, A, Barozzi, S, Parazzoli, D, Frittoli, E, Palamidessi, A, Panciera, T, Piccolo, S, Scita, G, Maiuri, P, Havas, K M, Zhou, Z W, Kumar, A, Bartek, J, Wang, Z Q, and Foiani, M
- Subjects
Cell Nucleus ,Mice, Knockout ,Cytoplasm ,Nuclear Envelope ,Science ,Neurogenesis ,[SDV]Life Sciences [q-bio] ,Brain ,Ataxia Telangiectasia Mutated Proteins ,Article ,Chromatin ,Cell invasion ,Actin Cytoskeleton ,Checkpoint signalling ,Animals ,lcsh:Q ,Stress, Mechanical ,Neoplasm Metastasis ,lcsh:Science ,Cytoskeleton ,DNA Damage - Abstract
ATR responds to mechanical stress at the nuclear envelope and mediates envelope-associated repair of aberrant topological DNA states. By combining microscopy, electron microscopic analysis, biophysical and in vivo models, we report that ATR-defective cells exhibit altered nuclear plasticity and YAP delocalization. When subjected to mechanical stress or undergoing interstitial migration, ATR-defective nuclei collapse accumulating nuclear envelope ruptures and perinuclear cGAS, which indicate loss of nuclear envelope integrity, and aberrant perinuclear chromatin status. ATR-defective cells also are defective in neuronal migration during development and in metastatic dissemination from circulating tumor cells. Our findings indicate that ATR ensures mechanical coupling of the cytoskeleton to the nuclear envelope and accompanying regulation of envelope-chromosome association. Thus the repertoire of ATR-regulated biological processes extends well beyond its canonical role in triggering biochemical implementation of the DNA damage response., The nucleus is a mechanically stiff organelle of the cell and the DNA damage response protein ATR can localize to the nuclear envelope upon mechanical stress. Here, the authors show that ATR may contribute to the integrity of the nuclear envelope and may play a role in cell migration.
- Published
- 2020
- Full Text
- View/download PDF
31. Exo1 Processes Stalled Replication Forks and Counteracts Fork Reversal in Checkpoint-Defective Cells
- Author
-
Ylli Doksani, Marco Foiani, Daniele Fachinetti, Jose' Sogo, Massimo Lopes, Cecilia Cotta-Ramusino, Chiara Lucca, University of Zurich, and Foiani, M
- Subjects
DNA Replication ,Genome instability ,Saccharomyces cerevisiae Proteins ,Genes, Fungal ,DNA, Single-Stranded ,Cell Cycle Proteins ,Saccharomyces cerevisiae ,Protein Serine-Threonine Kinases ,Biology ,medicine.disease_cause ,1307 Cell Biology ,chemistry.chemical_compound ,1312 Molecular Biology ,medicine ,Hydroxyurea ,Sister chromatids ,DNA, Fungal ,Molecular Biology ,Checkpoint Kinase 2 ,Mutation ,Cell Cycle ,10061 Institute of Molecular Cancer Research ,DNA replication ,Cell Biology ,Cell cycle ,Molecular biology ,Cell biology ,Microscopy, Electron ,Exodeoxyribonucleases ,chemistry ,570 Life sciences ,biology ,Fork (file system) ,DNA - Abstract
The replication checkpoint coordinates the cell cycle with DNA replication and recombination, preventing genome instability and cancer. The budding yeast Rad53 checkpoint kinase stabilizes stalled forks and replisome-fork complexes, thus preventing the accumulation of ss-DNA regions and reversed forks at collapsed forks. We searched for factors involved in the processing of stalled forks in HU-treated rad53 cells. Using the neutral-neutral two-dimensional electrophoresis technique (2D gel) and psoralen crosslinking combined with electron microscopy (EM), we found that the Exo1 exonuclease is recruited to stalled forks and, in rad53 mutants, counteracts reversed fork accumulation by generating ss-DNA intermediates. Hence, Exo1-mediated fork processing resembles the action of E. coli RecJ nuclease at damaged forks. Fork stability and replication restart are influenced by both DNA polymerase-fork association and Exo1-mediated processing. We suggest that Exo1 counteracts fork reversal by resecting newly synthesized chains and resolving the sister chromatid junctions that cause regression of collapsed forks.
- Published
- 2005
- Full Text
- View/download PDF
32. PP2A Controls Genome Integrity by Integrating Nutrient-Sensing and Metabolic Pathways with the DNA Damage Response
- Author
-
Elisa Ferrari, Walter Carotenuto, Rodrigo Bermejo, Marta Peretti, Corinne Cassani, Mario Varasi, Ghadeer Shubassi, Mohamed Elgendy, Chiara Lucca, Saverio Minucci, Christopher Bruhn, Marco Foiani, Maria Pia Longhese, Ferrari, E, Bruhn, C, Peretti, M, Cassani, C, Carotenuto, W, Elgendy, M, Shubassi, G, Lucca, C, Bermejo, R, Varasi, M, Minucci, S, Longhese, M, Foiani, M, Associazione Italiana per la Ricerca sul Cancro, Fondazione Telethon, and Ministero dell'Istruzione, dell'Università e della Ricerca
- Subjects
0301 basic medicine ,Methyltransferase ,Protein Methyltransferase ,DNA Repair ,Transcription Factor ,protein phosphatase PP2A ,Mutant ,Protein-Serine-Threonine Kinase ,DNA damage response ,environment and public health ,Ceramide ,0302 clinical medicine ,Gene Expression Regulation, Fungal ,Mec1-ATR ,Sirolimu ,Protein Phosphatase 2 ,DNA, Fungal ,chemistry.chemical_classification ,Intracellular Signaling Peptides and Proteins ,Cell biology ,Biochemistry ,Irc21 ,Genome, Fungal ,Cytochrome-B(5) Reductase ,Saccharomyces cerevisiae Protein ,Saccharomyces cerevisiae Proteins ,DNA damage ,Phosphatase ,Protein Kinase Inhibitor ,Metabolomic ,BIO/18 - GENETICA ,Saccharomyces cerevisiae ,Nutrient sensing ,Protein Serine-Threonine Kinases ,Biology ,Ceramides ,Article ,Genomic Instability ,03 medical and health sciences ,Genome stability ,Metabolomics ,Protein Methyltransferases ,Protein Kinase Inhibitors ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Sirolimus ,Cell Biology ,Protein phosphatase 2 ,TORC1 ,Enzyme Activation ,body regions ,Metabolic pathway ,Protein phosphatase PP2A ,Metabolism ,030104 developmental biology ,Enzyme ,chemistry ,Intracellular Signaling Peptides and Protein ,Rad53 ,Mutation ,Energy Metabolism ,metabolism ,genome stability ,030217 neurology & neurosurgery ,Transcription Factors ,DNA Damage - Abstract
Summary Mec1ATR mediates the DNA damage response (DDR), integrating chromosomal signals and mechanical stimuli. We show that the PP2A phosphatases, ceramide-activated enzymes, couple cell metabolism with the DDR. Using genomic screens, metabolic analysis, and genetic and pharmacological studies, we found that PP2A attenuates the DDR and that three metabolic circuits influence the DDR by modulating PP2A activity. Irc21, a putative cytochrome b5 reductase that promotes the condensation reaction generating dihydroceramides (DHCs), and Ppm1, a PP2A methyltransferase, counteract the DDR by activating PP2A; conversely, the nutrient-sensing TORC1-Tap42 axis sustains DDR activation by inhibiting PP2A. Loss-of-function mutations in IRC21, PPM1, and PP2A and hyperactive tap42 alleles rescue mec1 mutants. Ceramides synergize with rapamycin, a TORC1 inhibitor, in counteracting the DDR. Hence, PP2A integrates nutrient-sensing and metabolic pathways to attenuate the Mec1ATR response. Our observations imply that metabolic changes affect genome integrity and may help with exploiting therapeutic options and repositioning known drugs., Graphical Abstract, Highlights • PP2A counteracts the DNA damage response • Cytochrome b5-like Irc21 attenuates the DDR through ceramide-induced PP2A activation • The TORC1-Tap42 axis contributes to DDR activation through PP2A inhibition • The SAM-Ppm1 pathway attenuates the DDR by activating PP2A, Ferrari et al. provide a model wherein cytoplasmic metabolic pathways (the TORC1-Tap42, Ceramide-Irc21, and SAM-Ppm1 axes) converge on PP2A and PP2A-like phosphatases to modulate the nuclear DNA damage response. This reveals a connection between cell metabolism and genome stability surveillance mechanisms.
- Published
- 2017
- Full Text
- View/download PDF
33. Branch migrating sister chromatid junctions form at replication origins through Rad51/Rad52-independent mechanisms
- Author
-
Massimo Lopes, Marco Foiani, Giordano Liberi, Cecilia Cotta-Ramusino, University of Zurich, and Foiani, M
- Subjects
DNA Replication ,Saccharomyces cerevisiae Proteins ,Cell Cycle Proteins ,Replication Origin ,Saccharomyces cerevisiae ,Biology ,Chromatids ,Protein Serine-Threonine Kinases ,Origin of replication ,1307 Cell Biology ,Control of chromosome duplication ,1312 Molecular Biology ,Sister chromatids ,Replicon ,DNA, Fungal ,Molecular Biology ,Genetics ,Recombination, Genetic ,10061 Institute of Molecular Cancer Research ,DNA replication ,Cell Biology ,Rad52 DNA Repair and Recombination Protein ,Establishment of sister chromatid cohesion ,DNA-Binding Proteins ,Checkpoint Kinase 2 ,DNA Nucleotidyltransferases ,Origin recognition complex ,570 Life sciences ,biology ,Nucleic Acid Conformation ,Rad51 Recombinase ,Homologous recombination ,DNA Damage - Abstract
Cells overcome intra-S DNA damage and replication impediments by coupling chromosome replication to sister chromatid-mediated recombination and replication-bypass processes. Further, molecular junctions between replicated molecules have been suggested to assist sister chromatid cohesion until anaphase. Using two-dimensional gel electrophoresis, we have identified, in yeast cells, replication-dependent X-shaped molecules that appear during origin activation, branch migrate, and distribute along the replicon through a mechanism influenced by the rate of fork progression. Their formation is independent of Rad51- and Rad52-mediated homologous recombination events and is not affected by DNA damage or replication blocks. Further, in hydroxyurea-treated rad53 mutants, altered in the replication checkpoint, the branched molecules progressively degenerate and likely contribute to generate pathological structures. We suggest that cells couple sister chromatid tethering with replication initiation by generating specialized joint molecules resembling hemicatenanes: this process might prime cohesion and assist sister chromatid-mediated recombination and replication events.
- Published
- 2003
34. Fork reversal and ssDNA accumulation at stalled replication forks owing to checkpoint defects
- Author
-
Marco Foiani, José M. Sogo, Massimo Lopes, University of Zurich, and Foiani, M
- Subjects
Replication fork reversal ,Genome instability ,DNA Replication ,Saccharomyces cerevisiae Proteins ,DNA, Single-Stranded ,Cell Cycle Proteins ,Saccharomyces cerevisiae ,Biology ,Protein Serine-Threonine Kinases ,medicine.disease_cause ,Replication fork protection ,Replication fork protection complex ,Minichromosome maintenance ,Furocoumarins ,medicine ,Holliday junction ,Hydroxyurea ,Phosphorylation ,DNA, Fungal ,Genetics ,Recombination, Genetic ,1000 Multidisciplinary ,Mutation ,Multidisciplinary ,10061 Institute of Molecular Cancer Research ,Cell biology ,Nucleosomes ,DNA replication checkpoint ,Checkpoint Kinase 2 ,Microscopy, Electron ,Cross-Linking Reagents ,570 Life sciences ,biology ,Nucleic Acid Conformation - Abstract
Checkpoint-mediated control of replicating chromosomes is essential for preventing cancer. In yeast, Rad53 kinase protects stalled replication forks from pathological rearrangements. To characterize the mechanisms controlling fork integrity, we analyzed replication intermediates formed in response to replication blocks using electron microscopy. At the forks, wild-type cells accumulate short single-stranded regions, which likely causes checkpoint activation, whereas rad53 mutants exhibit extensive single-stranded gaps and hemi-replicated intermediates, consistent with a lagging-strand synthesis defect. Further, rad53 cells accumulate Holliday junctions through fork reversal. We speculate that, in checkpoint mutants, abnormal replication intermediates begin to form because of uncoordinated replication and are further processed by unscheduled recombination pathways, causing genome instability.
- Published
- 2002
35. PDXK mutations cause polyneuropathy responsive to pyridoxal 5′‐phosphate supplementation
- Author
-
Chelban, Viorica, Wilson, Matthew P., Warman Chardon, Jodi, Vandrovcova, Jana, Zanetti, M. Natalia, Zamba‐Papanicolaou, Eleni, Efthymiou, Stephanie, Pope, Simon, Conte, Maria R., Abis, Giancarlo, Liu, Yo‐Tsen, Tribollet, Eloise, Haridy, Nourelhoda A., Botía, Juan A., Ryten, Mina, Nicolaou, Paschalis, Minaidou, Anna, Christodoulou, Kyproula, Kernohan, Kristin D., Eaton, Alison, Osmond, Matthew, Ito, Yoko, Bourque, Pierre, Jepson, James E. C., Bello, Oscar, Bremner, Fion, Cordivari, Carla, Reilly, Mary M., Foiani, Martha, Heslegrave, Amanda, Zetterberg, Henrik, Heales, Simon J. R., Wood, Nicholas W., Rothman, James E., Boycott, Kym M., Mills, Philippa B., Clayton, Peter T., Houlden, Henry, Kriouile, Yamna, Khorassani, Mohamed El, Aguennouz, Mhammed, Groppa, Stanislav, Marinova Karashova, Blagovesta, Van Maldergem, Lionel, Nachbauer, Wolfgang, Boesch, Sylvia, Arning, Larissa, Timmann, Dagmar, Cormand, Bru, Pérez‐Dueñas, Belen, Di Rosa, Gabriella, Goraya, Jatinder S., Sultan, Tipu, Mine, Jun, Avdjieva, Daniela, Kathom, Hadil, Tincheva, Radka, Banu, Selina, Pineda‐Marfa, Mercedes, Veggiotti, Pierangelo, Ferrari, Michel D., van den Maagdenberg, Arn M J M, Verrotti, Alberto, Marseglia, Giangluigi, Savasta, Salvatore, García‐Silva, Mayte, Ruiz, Alfons Macaya, Garavaglia, Barbara, Borgione, Eugenia, Portaro, Simona, Sanchez, Benigno Monteagudo, Boles, Richard, Papacostas, Savvas, Vikelis, Michail, Rothman, James, Giunti, Paola, Salpietro, Vincenzo, Oconnor, Emer, Kullmann, Dimitri, Kaiyrzhanov, Rauan, Sullivan, Roisin, Khan, Alaa Matooq, Yau, Wai Yan, Hostettler, Isabel, Papanicolaou, Eleni Zamba, Dardiotis, Efthymios, Maqbool, Shazia, Ibrahim, Shahnaz, Kirmani, Salman, Rana, Nuzhat Noureen, Atawneh, Osama, Lim, Shen‐Yang, Shaikh, Farooq, Koutsis, George, Breza, Marianthi, Mangano, Salvatore, Scuderi, Carmela, Morello, Giovanna, Stojkovic, Tanya, Torti, Erin, Zollo, Massimi, Heimer, Gali, Dauvilliers, Yves A., Striano, Pasquale, Al‐Khawaja, Issam, Al‐Mutairi, Fuad, Alkuraya, Fowzan S, Sherifa, Hamed, Rizig, Mie, Okubadejo, Njideka U., Ojo, Oluwadamilola O., Oshinaike, Olajumoke O., Wahab, Kolawole, Bello, Abiodun H., Abubakar, Sanni, Obiabo, Yahaya, Nwazor, Ernest, Ekenze, Oluchi, Williams, Uduak, Iyagba, Alagoma, Taiwo, Lolade, Komolafe, Morenikeji, Oguntunde, Olapeju, Pchelina, Sofya, Senkevich, Konstantin, Haridy, Nourelhoda, Shashkin, Chingiz, Zharkynbekova, Nazira, Koneyev, Kairgali, Manizha, Ganieva, Isrofilov, Maksud, Guliyeva, Ulviyya, Salayev, Kamran, Khachatryan, Samson, Rossi, Salvatore, Silvestri, Gabriella, Bourinaris, Thomas, Xiromerisiou, Georgia, Fidani, Liana, Spanaki, Cleanthe, Tucci, Arianna, University College London Hospitals (UCLH), Université d'Ottawa [Ontario] (uOttawa), King‘s College London, University College of London [London] (UCL), University of Cyprus [Nicosia], UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, Queen Square, London, Institute of Psychiatry, Psychology & Neuroscience, King's College London, National Yang Ming University (NYMU), Department of Information and Communications Engineering [Murcia], Universidad de Murcia, Guy's Hospital [London], Cyprus Institute of Neurology and Genetics, University of Ottawa [Ottawa], The Ottawa Hospital, University of British Columbia (UBC), Ottawa Hospital Research Institute [Ottawa] (OHRI), Institute of Neurology, Queen Square, London, Sahlgrenska University Hospital, Biochemistry, Endocrinology and Metabolism Unit, Institute of Child Health, Laboratoire Chrono-environnement - CNRS - UBFC (UMR 6249) (LCE), Centre National de la Recherche Scientifique (CNRS)-Université de Franche-Comté (UFC), Université Bourgogne Franche-Comté [COMUE] (UBFC)-Université Bourgogne Franche-Comté [COMUE] (UBFC), Department of Human Genetics, Ruhr University Bochum (RUB), Universitat de Barcelona (UB), Department of Medical and Surgical Pediatrics, University Hospital, Fondazione, Departments of Human Genetics & Neurology, Leiden University Medical Center (LUMC), University of Laquila, Fondazione IRCCS Istituto Neurologico 'Carlo Besta', Yale University School of Medicine, Department of Microbiology, Università degli studi di Catania [Catania], CHU Pitié-Salpêtrière [AP-HP], Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Gene Dx, Partenaires INRAE, Tel Aviv University Sackler School of Medicine [Tel Aviv, Israël], Neuropsychiatrie : recherche épidémiologique et clinique (PSNREC), Université Montpellier 1 (UM1)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier), Universita degli studi di Genova, Fondazione 'Policlinico Universitario A. Gemelli' [Rome], UCL Institute of neurology, UCL Institute of Neurology, Chelban V., Wilson M.P., Warman Chardon J., Vandrovcova J., Zanetti M.N., Zamba-Papanicolaou E., Efthymiou S., Pope S., Conte M.R., Abis G., Liu Y.-T., Tribollet E., Haridy N.A., Botia J.A., Ryten M., Nicolaou P., Minaidou A., Christodoulou K., Kernohan K.D., Eaton A., Osmond M., Ito Y., Bourque P., Jepson J.E.C., Bello O., Bremner F., Cordivari C., Reilly M.M., Foiani M., Heslegrave A., Zetterberg H., Heales S.J.R., Wood N.W., Rothman J.E., Boycott K.M., Mills P.B., Clayton P.T., Houlden H., Kriouile Y., Khorassani M.E., Aguennouz M., Groppa S., Marinova Karashova B., Van Maldergem L., Nachbauer W., Boesch S., Arning L., Timmann D., Cormand B., Perez-Duenas B., Di Rosa G., Goraya J.S., Sultan T., Mine J., Avdjieva D., Kathom H., Tincheva R., Banu S., Pineda-Marfa M., Veggiotti P., Ferrari M.D., van den Maagdenberg A.M.J.M., Verrotti A., Marseglia G., Savasta S., Garcia-Silva M., Ruiz A.M., Garavaglia B., Borgione E., Portaro S., Sanchez B.M., Boles R., Papacostas S., Vikelis M., Rothman J., Giunti P., Salpietro V., Oconnor E., Kullmann D., Kaiyrzhanov R., Sullivan R., Khan A.M., Yau W.Y., Hostettler I., Papanicolaou E.Z., Dardiotis E., Maqbool S., Ibrahim S., Kirmani S., Rana N.N., Atawneh O., Lim S.-Y., Shaikh F., Koutsis G., Breza M., Mangano S., Scuderi C., Morello G., Stojkovic T., Torti E., Zollo M., Heimer G., Dauvilliers Y.A., Striano P., Al-Khawaja I., Al-Mutairi F., Alkuraya F.S., Sherifa H., Rizig M., Okubadejo N.U., Ojo O.O., Oshinaike O.O., Wahab K., Bello A.H., Abubakar S., Obiabo Y., Nwazor E., Ekenze O., Williams U., Iyagba A., Taiwo L., Komolafe M., Oguntunde O., Pchelina S., Senkevich K., Haridy N., Shashkin C., Zharkynbekova N., Koneyev K., Manizha G., Isrofilov M., Guliyeva U., Salayev K., Khachatryan S., Rossi S., Silvestri G., Bourinaris T., Xiromerisiou G., Fidani L., Spanaki C., and Tucci A.
- Subjects
0301 basic medicine ,Male ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,LOCAL TRANSLATION ,Medizin ,medicine.disease_cause ,DISEASE ,chemistry.chemical_compound ,0302 clinical medicine ,polineuropathy ,Cinètica enzimàtica ,Gene Regulatory Networks ,Pyridoxal phosphate ,Child ,Pyridoxal Kinase ,Adenosine triphosphate (ATP) ,Research Articles ,Aged, 80 and over ,Mutation ,Gene Regulatory Network ,PLASMA ,Autosomal recessive axonal polyneuropathy ,Disease gene identification ,Pyridoxal kinase ,3. Good health ,Settore MED/26 - NEUROLOGIA ,Neuropaties perifèriques ,Treatment Outcome ,Polyneuropathie ,Neurology ,Child, Preschool ,Pyridoxal Phosphate ,RELIABILITY ,Vitamin B Complex ,Female ,Life Sciences & Biomedicine ,Polyneuropathy ,Human ,Research Article ,Adult ,Adolescent ,PDXK ,Clinical Neurology ,CHARCOT-MARIE-TOOTH ,CHARCOT-MARIE-TOOTH, CMT NEUROPATHY SCORE, LOCAL TRANSLATION, DISEASE, RELIABILITY ,MECHANISMS, DISCOVERY, FRAMEWORK, KINASE, PLASMA ,MECHANISMS ,03 medical and health sciences ,Polyneuropathies ,Atrophy ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,KINASE ,medicine ,Humans ,CMT NEUROPATHY SCORE ,PDXK mutations ,Pyridoxal ,Dietary Supplement ,Aged ,Peripheral neuropathies ,Science & Technology ,[SCCO.NEUR]Cognitive science/Neuroscience ,Enzyme kinetics ,Neurosciences ,FRAMEWORK ,medicine.disease ,Molecular biology ,030104 developmental biology ,chemistry ,DISCOVERY ,Dietary Supplements ,Neurosciences & Neurology ,Neurology (clinical) ,Adenosine triphosphate ,030217 neurology & neurosurgery - Abstract
OBJECTIVE: To identify disease-causing variants in autosomal recessive axonal polyneuropathy with optic atrophy and provide targeted replacement therapy. METHODS: We performed genome-wide sequencing, homozygosity mapping, and segregation analysis for novel disease-causing gene discovery. We used circular dichroism to show secondary structure changes and isothermal titration calorimetry to investigate the impact of variants on adenosine triphosphate (ATP) binding. Pathogenicity was further supported by enzymatic assays and mass spectroscopy on recombinant protein, patient-derived fibroblasts, plasma, and erythrocytes. Response to supplementation was measured with clinical validated rating scales, electrophysiology, and biochemical quantification. RESULTS: We identified biallelic mutations in PDXK in 5 individuals from 2 unrelated families with primary axonal polyneuropathy and optic atrophy. The natural history of this disorder suggests that untreated, affected individuals become wheelchair-bound and blind. We identified conformational rearrangement in the mutant enzyme around the ATP-binding pocket. Low PDXK ATP binding resulted in decreased erythrocyte PDXK activity and low pyridoxal 5'-phosphate (PLP) concentrations. We rescued the clinical and biochemical profile with PLP supplementation in 1 family, improvement in power, pain, and fatigue contributing to patients regaining their ability to walk independently during the first year of PLP normalization. INTERPRETATION: We show that mutations in PDXK cause autosomal recessive axonal peripheral polyneuropathy leading to disease via reduced PDXK enzymatic activity and low PLP. We show that the biochemical profile can be rescued with PLP supplementation associated with clinical improvement. As B6 is a cofactor in diverse essential biological pathways, our findings may have direct implications for neuropathies of unknown etiology characterized by reduced PLP levels. ANN NEUROL 2019;86:225-240. ispartof: ANNALS OF NEUROLOGY vol:86 issue:2 pages:225-240 ispartof: location:United States status: published
- Full Text
- View/download PDF
36. The integrated stress response drives MET oncogene overexpression in cancers.
- Author
-
Cerqua M, Foiani M, Boccaccio C, Comoglio PM, and Altintas DM
- Abstract
Cancer cells rely on invasive growth to survive in a hostile microenvironment; this growth is characterised by interconnected processes such as epithelial-to-mesenchymal transition and migration. A master regulator of these events is the MET oncogene, which is overexpressed in the majority of cancers; however, since mutations in the MET oncogene are seen only rarely in cancers and are relatively infrequent, the mechanisms that cause this widespread MET overexpression remain obscure. Here, we show that the 5' untranslated region (5'UTR) of MET mRNA harbours two functional stress-responsive elements, conferring translational regulation by the integrated stress response (ISR), regulated by phosphorylation of eukaryotic translation initiation factor 2 alpha (eIF2α) at serine 52. ISR activation by serum starvation, leucine deprivation, hypoxia, irradiation, thapsigargin or gemcitabine is followed by MET protein overexpression. We mechanistically link MET translation to the ISR by (i) mutation of the two uORFs within the MET 5'UTR, (ii) CRISPR/Cas9-mediated mutation of eIF2α (S52A), or (iii) the application of ISR pathway inhibitors. All of these interventions reduce stress-induced MET overexpression. Finally, we show that blocking stress-induced MET translation blunts MET-dependent invasive growth. These findings indicate that upregulation of the MET oncogene is a functional requirement linking integrated stress response to cancer progression., Competing Interests: Disclosure and competing interests statement. The authors declare no competing interests., (© 2024. The Author(s).)
- Published
- 2025
- Full Text
- View/download PDF
37. A multiparametric screen uncovers FDA-approved small molecules that potentiate the nuclear mechano-dysfunctions in ATR-defective cells.
- Author
-
Cera MR, Bastianello G, Purushothaman D, Andronache A, Ascione F, Robusto M, Fagà G, Pasi M, Meroni G, Li Q, Choudhary R, Varasi M, Foiani M, and Mercurio C
- Subjects
- Humans, HeLa Cells, Small Molecule Libraries pharmacology, Cell Line, Tumor, Morpholines pharmacology, Cell Nucleus metabolism, Cell Nucleus drug effects, Pyrimidines pharmacology, Nuclear Envelope metabolism, Nuclear Envelope drug effects, Indoles, Membrane Proteins, Sulfonamides, Ataxia Telangiectasia Mutated Proteins metabolism
- Abstract
Targeting nuclear mechanics is emerging as a promising therapeutic strategy for sensitizing cancer cells to immunotherapy. Inhibition of the mechano-sensory kinase ATR leads to mechanical vulnerability of cancer cells, causing nuclear envelope softness and collapse and activation of the cGAS-STING-mediated innate immune response. Finding novel compounds that interfere with the non-canonical role of ATR in controlling nuclear mechanics presents an intriguing therapeutic opportunity. We carried out a multiparametric high-content screen to identify small molecules that affect nuclear envelope shape and to uncover novel players that could either ameliorate or further compromise the nuclear mechanical abnormalities of ATR-defective cells. The screen was performed in HeLa cells genetically depleted for ATR. Candidate hits were also tested in combination with the chemical inhibition of ATR by AZD6738, and their efficacy was further validated in the triple-negative breast cancer cell lines BT549 and HCC1937. We show that those compounds enhancing the abnormal nuclear shape of ATR-defective cells also synergize with AZD6738 to boost the expression of interferon-stimulated genes, highlighting the power of multiparametric screens to identify novel combined therapeutic interventions targeting nuclear mechanics for cancer immunotherapy., Competing Interests: Declarations. Competing interests: The authors declare no competing interests., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
38. Mechanical stress during confined migration causes aberrant mitoses and c-MYC amplification.
- Author
-
Bastianello G, Kidiyoor GR, Lowndes C, Li Q, Bonnal R, Godwin J, Iannelli F, Drufuca L, Bason R, Orsenigo F, Parazzoli D, Pavani M, Cancila V, Piccolo S, Scita G, Ciliberto A, Tripodo C, Pagani M, and Foiani M
- Subjects
- Humans, Animals, Cell Line, Tumor, Mice, Mitosis genetics, Chromosomal Instability, Gene Expression Regulation, Neoplastic, Neoplasms genetics, Neoplasms pathology, Neoplasms metabolism, Cell Movement genetics, Proto-Oncogene Proteins c-myc metabolism, Proto-Oncogene Proteins c-myc genetics, Stress, Mechanical, Gene Amplification
- Abstract
Confined cell migration hampers genome integrity and activates the ATR and ATM mechano-transduction pathways. We investigated whether the mechanical stress generated by metastatic interstitial migration contributes to the enhanced chromosomal instability observed in metastatic tumor cells. We employed live cell imaging, micro-fluidic approaches, and scRNA-seq to follow the fate of tumor cells experiencing confined migration. We found that, despite functional ATR, ATM, and spindle assembly checkpoint (SAC) pathways, tumor cells dividing across constriction frequently exhibited altered spindle pole organization, chromosome mis-segregations, micronuclei formation, chromosome fragility, high gene copy number variation, and transcriptional de-regulation and up-regulation of c-MYC oncogenic transcriptional signature via c-MYC locus amplifications. In vivo tumor settings showed that malignant cells populating metastatic foci or infiltrating the interstitial stroma gave rise to cells expressing high levels of c-MYC. Altogether, our data suggest that mechanical stress during metastatic migration contributes to override the checkpoint controls and boosts genotoxic and oncogenic events. Our findings may explain why cancer aneuploidy often does not correlate with mutations in SAC genes and why c-MYC amplification is strongly linked to metastatic tumors., Competing Interests: Competing interests statement:M.P. is of the board of Directors and stakeholder of CheckmAb s.r.l. and is a recipient of grants under a research agreement with Bristol-Myers Squibb and Macomics.
- Published
- 2024
- Full Text
- View/download PDF
39. Sch9 S6K controls DNA repair and DNA damage response efficiency in aging cells.
- Author
-
Lucca C, Ferrari E, Shubassi G, Ajazi A, Choudhary R, Bruhn C, Matafora V, Bachi A, and Foiani M
- Subjects
- DNA Damage, Intracellular Signaling Peptides and Proteins metabolism, Intracellular Signaling Peptides and Proteins genetics, Phosphorylation, Protein Phosphatase 2 metabolism, Saccharomyces cerevisiae cytology, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae physiology, Cellular Senescence, DNA Repair, Protein Serine-Threonine Kinases metabolism, Protein Serine-Threonine Kinases genetics, Saccharomyces cerevisiae Proteins metabolism, Saccharomyces cerevisiae Proteins genetics
- Abstract
Survival from UV-induced DNA lesions relies on nucleotide excision repair (NER) and the Mec1
ATR DNA damage response (DDR). We study DDR and NER in aging cells and find that old cells struggle to repair DNA and activate Mec1ATR . We employ pharmacological and genetic approaches to rescue DDR and NER during aging. Conditions activating Snf1AMPK rescue DDR functionality, but not NER, while inhibition of the TORC1-Sch9S6K axis restores NER and enhances DDR by tuning PP2A activity, specifically in aging cells. Age-related repair deficiency depends on Snf1AMPK -mediated phosphorylation of Sch9S6K on Ser160 and Ser163. PP2A activity in old cells is detrimental for DDR and influences NER by modulating Snf1AMPK and Sch9S6K . Hence, the DDR and repair pathways in aging cells are influenced by the metabolic tuning of opposing AMPK and TORC1 networks and by PP2A activity. Specific Sch9S6K phospho-isoforms control DDR and NER efficiency, specifically during aging., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
40. Cell stretching activates an ATM mechano-transduction pathway that remodels cytoskeleton and chromatin.
- Author
-
Bastianello G, Porcella G, Beznoussenko GV, Kidiyoor G, Ascione F, Li Q, Cattaneo A, Matafora V, Disanza A, Quarto M, Mironov AA, Oldani A, Barozzi S, Bachi A, Costanzo V, Scita G, and Foiani M
- Subjects
- Humans, Cell Cycle Proteins metabolism, Tumor Suppressor Proteins metabolism, Ataxia Telangiectasia Mutated Proteins metabolism, Chromatin metabolism, Reactive Oxygen Species metabolism, Proteomics, DNA-Binding Proteins metabolism, Phosphorylation, DNA Damage, Cytoskeleton metabolism, Protein Serine-Threonine Kinases metabolism, Ataxia Telangiectasia
- Abstract
Ataxia telangiectasia mutated (ATM) and ataxia telangiectasia and Rad3-related (ATR) DNA damage response (DDR) kinases contain elastic domains. ATM also responds to reactive oxygen species (ROS) and ATR to nuclear mechanical stress. Mre11 mediates ATM activation following DNA damage; ATM mutations cause ataxia telangiectasia (A-T). Here, using in vivo imaging, electron microscopy, proteomic, and mechano-biology approaches, we study how ATM responds to mechanical stress. We report that cytoskeleton and ROS, but not Mre11, mediate ATM activation following cell deformation. ATM deficiency causes hyper-stiffness, stress fiber accumulation, Yes-associated protein (YAP) nuclear enrichment, plasma and nuclear membrane alterations during interstitial migration, and H3 hyper-methylation. ATM locates to the actin cytoskeleton and, following cytoskeleton stress, promotes phosphorylation of key cytoskeleton and chromatin regulators. Our data contribute to explain some clinical features of patients with A-T and pinpoint the existence of an integrated mechano-response in which ATM and ATR have distinct roles unrelated to their canonical DDR functions., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
41. Sen1 and Rrm3 ensure permissive topological conditions for replication termination.
- Author
-
Choudhary R, Niska-Blakie J, Adhil M, Liberi G, Achar YJ, Giannattasio M, and Foiani M
- Subjects
- DNA, DNA Replication, DNA Topoisomerases, Type II metabolism, DNA Helicases genetics, DNA Helicases metabolism, RNA Helicases genetics, RNA Helicases metabolism, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins metabolism
- Abstract
Replication forks terminate at TERs and telomeres. Forks that converge or encounter transcription generate topological stress. Combining genetics, genomics, and transmission electron microscopy, we find that Rrm3
hPif1 and Sen1hSenataxin helicases assist termination at TERs; Sen1 specifically acts at telomeres. rrm3 and sen1 genetically interact and fail to terminate replication, exhibiting fragility at termination zones (TERs) and telomeres. sen1rrm3 accumulates RNA-DNA hybrids and X-shaped gapped or reversed converging forks at TERs; sen1, but not rrm3, builds up RNA polymerase II (RNPII) at TERs and telomeres. Rrm3 and Sen1 restrain Top1 and Top2 activities, preventing toxic accumulation of positive supercoil at TERs and telomeres. We suggest that Rrm3 and Sen1 coordinate the activities of Top1 and Top2 when forks encounter transcription head on or codirectionally, respectively, thus preventing the slowing down of DNA and RNA polymerases. Hence Rrm3 and Sen1 are indispensable to generate permissive topological conditions for replication termination., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
42. Endogenous PP2A inhibitor CIP2A degradation by chaperone-mediated autophagy contributes to the antitumor effect of mitochondrial complex I inhibition.
- Author
-
Cazzoli R, Romeo F, Pallavicini I, Peri S, Romanenghi M, Pérez-Valencia JA, Hagag E, Ferrucci F, Elgendy M, Vittorio O, Pece S, Foiani M, Westermarck J, and Minucci S
- Subjects
- Humans, Autoantigens metabolism, Cell Line, Tumor, Energy Metabolism, Oxidative Phosphorylation, Protein Phosphatase 2 antagonists & inhibitors, Protein Phosphatase 2 metabolism, Signal Transduction, Chaperone-Mediated Autophagy, Neoplasms pathology, Electron Transport Complex I antagonists & inhibitors
- Abstract
Combined inhibition of oxidative phosphorylation (OXPHOS) and glycolysis has been shown to activate a PP2A-dependent signaling pathway, leading to tumor cell death. Here, we analyze highly selective mitochondrial complex I or III inhibitors in vitro and in vivo to elucidate the molecular mechanisms leading to cell death following OXPHOS inhibition. We show that IACS-010759 treatment (complex I inhibitor) induces a reactive oxygen species (ROS)-dependent dissociation of CIP2A from PP2A, leading to its destabilization and degradation through chaperone-mediated autophagy. Mitochondrial complex III inhibition has analogous effects. We establish that activation of the PP2A holoenzyme containing B56δ regulatory subunit selectively mediates tumor cell death, while the arrest in proliferation that is observed upon IACS-010759 treatment does not depend on the PP2A-B56δ complex. These studies provide a molecular characterization of the events subsequent to the alteration of critical bioenergetic pathways and help to refine clinical studies aimed to exploit metabolic vulnerabilities of tumor cells., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
43. Tau filaments are tethered within brain extracellular vesicles in Alzheimer's disease.
- Author
-
Fowler SL, Behr TS, Turkes E, Cauhy PM, Foiani MS, Schaler A, Crowley G, Bez S, Ficulle E, Tsefou E, O'Brien DP, Fischer R, Geary B, Gaur P, Miller C, D'Acunzo P, Levy E, Duff KE, and Ryskeldi-Falcon B
- Abstract
The abnormal assembly of tau protein in neurons is the pathological hallmark of multiple neurodegenerative diseases, including Alzheimer's disease (AD). In addition, assembled tau associates with extracellular vesicles (EVs) in the central nervous system of patients with AD, which is linked to its clearance and prion-like propagation between neurons. However, the identities of the assembled tau species and the EVs, as well as how they associate, are not known. Here, we combined quantitative mass spectrometry, cryo-electron tomography and single-particle cryo-electron microscopy to study brain EVs from AD patients. We found filaments of truncated tau enclosed within EVs enriched in endo-lysosomal proteins. We observed multiple filament interactions, including with molecules that tethered filaments to the EV limiting membrane, suggesting selective packaging. Our findings will guide studies into the molecular mechanisms of EV-mediated secretion of assembled tau and inform the targeting of EV-associated tau as potential therapeutic and biomarker strategies for AD.
- Published
- 2023
- Full Text
- View/download PDF
44. Cancer cell histone density links global histone acetylation, mitochondrial proteome and histone acetylase inhibitor sensitivity.
- Author
-
Bruhn C, Bastianello G, and Foiani M
- Subjects
- Acetylation, Chromatin, Histone Acetyltransferases, Proteome, Histones, Neoplasms
- Abstract
Chromatin metabolism is frequently altered in cancer cells and facilitates cancer development. While cancer cells produce large amounts of histones, the protein component of chromatin packaging, during replication, the potential impact of histone density on cancer biology has not been studied systematically. Here, we show that altered histone density affects global histone acetylation, histone deactylase inhibitor sensitivity and altered mitochondrial proteome composition. We present estimates of nuclear histone densities in 373 cancer cell lines, based on Cancer Cell Line Encyclopedia data, and we show that a known histone regulator, HMGB1, is linked to histone density aberrations in many cancer cell lines. We further identify an E3 ubiquitin ligase interactor, DCAF6, and a mitochondrial respiratory chain assembly factor, CHCHD4, as histone modulators. As systematic characterization of histone density aberrations in cancer cell lines, this study provides approaches and resources to investigate the impact of histone density on cancer biology., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
45. YAP/TAZ activity in stromal cells prevents ageing by controlling cGAS-STING.
- Author
-
Sladitschek-Martens HL, Guarnieri A, Brumana G, Zanconato F, Battilana G, Xiccato RL, Panciera T, Forcato M, Bicciato S, Guzzardo V, Fassan M, Ulliana L, Gandin A, Tripodo C, Foiani M, Brusatin G, Cordenonsi M, and Piccolo S
- Subjects
- Actin-Related Protein 2 metabolism, Cellular Senescence, Extracellular Matrix, Healthy Aging, Immunity, Innate, Lamin Type B metabolism, Mechanotransduction, Cellular genetics, Nuclear Envelope metabolism, Signal Transduction, Aging metabolism, Membrane Proteins metabolism, Nucleotidyltransferases metabolism, Stromal Cells metabolism, Transcriptional Coactivator with PDZ-Binding Motif Proteins antagonists & inhibitors, Transcriptional Coactivator with PDZ-Binding Motif Proteins metabolism, YAP-Signaling Proteins antagonists & inhibitors, YAP-Signaling Proteins metabolism
- Abstract
Ageing is intimately connected to the induction of cell senescence
1,2 , but why this is so remains poorly understood. A key challenge is the identification of pathways that normally suppress senescence, are lost during ageing and are functionally relevant to oppose ageing3 . Here we connected the structural and functional decline of ageing tissues to attenuated function of the master effectors of cellular mechanosignalling YAP and TAZ. YAP/TAZ activity declines during physiological ageing in stromal cells, and mimicking such decline through genetic inactivation of YAP/TAZ in these cells leads to accelerated ageing. Conversely, sustaining YAP function rejuvenates old cells and opposes the emergence of ageing-related traits associated with either physiological ageing or accelerated ageing triggered by a mechano-defective extracellular matrix. Ageing traits induced by inactivation of YAP/TAZ are preceded by induction of tissue senescence. This occurs because YAP/TAZ mechanotransduction suppresses cGAS-STING signalling, to the extent that inhibition of STING prevents tissue senescence and premature ageing-related tissue degeneration after YAP/TAZ inactivation. Mechanistically, YAP/TAZ-mediated control of cGAS-STING signalling relies on the unexpected role of YAP/TAZ in preserving nuclear envelope integrity, at least in part through direct transcriptional regulation of lamin B1 and ACTR2, the latter of which is involved in building the peri-nuclear actin cap. The findings demonstrate that declining YAP/TAZ mechanotransduction drives ageing by unleashing cGAS-STING signalling, a pillar of innate immunity. Thus, sustaining YAP/TAZ mechanosignalling or inhibiting STING may represent promising approaches for limiting senescence-associated inflammation and improving healthy ageing., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
46. Pressure Overload Activates DNA-Damage Response in Cardiac Stromal Cells: A Novel Mechanism Behind Heart Failure With Preserved Ejection Fraction?
- Author
-
Stadiotti I, Santoro R, Scopece A, Pirola S, Guarino A, Polvani G, Maione AS, Ascione F, Li Q, Delia D, Foiani M, Pompilio G, and Sommariva E
- Abstract
Heart failure with preserved ejection fraction (HFpEF) is a heterogeneous syndrome characterized by impaired left ventricular (LV) diastolic function, with normal LV ejection fraction. Aortic valve stenosis can cause an HFpEF-like syndrome by inducing sustained pressure overload (PO) and cardiac remodeling, as cardiomyocyte (CM) hypertrophy and fibrotic matrix deposition. Recently, in vivo studies linked PO maladaptive myocardial changes and DNA damage response (DDR) activation: DDR-persistent activation contributes to mouse CM hypertrophy and inflammation, promoting tissue remodeling, and HF. Despite the wide acknowledgment of the pivotal role of the stromal compartment in the fibrotic response to PO, the possible effects of DDR-persistent activation in cardiac stromal cell (C-MSC) are still unknown. Finally, this novel mechanism was not verified in human samples. This study aims to unravel the effects of PO-induced DDR on human C-MSC phenotypes. Human LV septum samples collected from severe aortic stenosis with HFpEF-like syndrome patients undergoing aortic valve surgery and healthy controls (HCs) were used both for histological tissue analyses and C-MSC isolation. PO-induced mechanical stimuli were simulated in vitro by cyclic unidirectional stretch. Interestingly, HFpEF tissue samples revealed DNA damage both in CM and C-MSC. DDR-activation markers γH2AX, pCHK1, and pCHK2 were expressed at higher levels in HFpEF total tissue than in HC. Primary C-MSC isolated from HFpEF and HC subjects and expanded in vitro confirmed the increased γH2AX and phosphorylated checkpoint protein expression, suggesting a persistent DDR response, in parallel with a higher expression of pro-fibrotic and pro-inflammatory factors respect to HC cells, hinting to a DDR-driven remodeling of HFpEF C-MSC. Pressure overload was simulated in vitro , and persistent activation of the CHK1 axis was induced in response to in vitro mechanical stretching, which also increased C-MSC secreted pro-inflammatory and pro-fibrotic molecules. Finally, fibrosis markers were reverted by the treatment with a CHK1/ATR pathway inhibitor, confirming a cause-effect relationship. In conclusion we demonstrated that, in severe aortic stenosis with HFpEF-like syndrome patients, PO induces DDR-persistent activation not only in CM but also in C-MSC. In C-MSC, DDR activation leads to inflammation and fibrosis, which can be prevented by specific DDR targeting., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Stadiotti, Santoro, Scopece, Pirola, Guarino, Polvani, Maione, Ascione, Li, Delia, Foiani, Pompilio and Sommariva.)
- Published
- 2022
- Full Text
- View/download PDF
47. The transcription factor PREP1(PKNOX1) regulates nuclear stiffness, the expression of LINC complex proteins and mechanotransduction.
- Author
-
Purushothaman D, Bianchi LF, Penkov D, Poli A, Li Q, Vermezovic J, Pramotton FM, Choudhary R, Pennacchio FA, Sommariva E, Foiani M, Gauthier N, Maiuri P, and Blasi F
- Subjects
- Mechanotransduction, Cellular physiology, Nuclear Envelope metabolism, Transcriptional Coactivator with PDZ-Binding Motif Proteins, YAP-Signaling Proteins, Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Transcription Factors genetics, Transcription Factors metabolism
- Abstract
Mechanosignaling, initiated by extracellular forces and propagated through the intracellular cytoskeletal network, triggers signaling cascades employed in processes as embryogenesis, tissue maintenance and disease development. While signal transduction by transcription factors occurs downstream of cellular mechanosensing, little is known about the cell intrinsic mechanisms that can regulate mechanosignaling. Here we show that transcription factor PREP1 (PKNOX1) regulates the stiffness of the nucleus, the expression of LINC complex proteins and mechanotransduction of YAP-TAZ. PREP1 depletion upsets the nuclear membrane protein stoichiometry and renders nuclei soft. Intriguingly, these cells display fortified actomyosin network with bigger focal adhesion complexes resulting in greater traction forces at the substratum. Despite the high traction, YAP-TAZ translocation is impaired indicating disrupted mechanotransduction. Our data demonstrate mechanosignaling upstream of YAP-TAZ and suggest the existence of a transcriptional mechanism actively regulating nuclear membrane homeostasis and signal transduction through the active engagement/disengagement of the cell from the extracellular matrix., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
48. Vps30/Atg6/BECN1 at the crossroads between cell metabolism and DNA damage response.
- Author
-
Ajazi A and Foiani M
- Subjects
- Autophagy-Related Proteins, Beclin-1, DNA Damage, Mechanistic Target of Rapamycin Complex 1, Amino Acids, Autophagy physiology
- Abstract
Several cytotoxic agents used in cancer therapy cause DNA damage and replication stress. Understanding the metabolic determinants of the cell response to replication stress-inducing agents could have relevant implications for cancer treatment. In a recent study, we showed that cell survival during replication stress is influenced by the availability of amino acids, as well as by TORC1 and Gcn2-mediated amino acid sensing pathways. Amino acid starvation, or TORC1 inhibition, sensitizes cells to replication stress conditions, whereas Gcn2 ablation promotes cell survival by stimulating protein synthesis. The Vps34-Vps15-Vps30/Atg6/BECN1-Vps38/UVRAG phosphatidylinositol-3-phosphate (PtdIns3P) complex at the endosomes sets the balance between survival and death signals during replication stress and amino acid starvation. The Vps34-Vps15-Vps30/Atg6/BECN1-Vps38/UVRAG axis promotes the degradation of amino acid transporters, thus sensitizing cells to amino acid starvation, while Vps34-Vps15-Vps30/Atg6/BECN1-Vps38/UVRAG inactivation promotes cell survival by enabling synthesis of stress response proteins mediating survival under replication stress conditions. Our study unravels an autophagy-independent mechanism through which Vps34-Vps30/Atg6/BECN1 promotes lethal events during replication stress.
- Published
- 2022
- Full Text
- View/download PDF
49. The ESCRT machinery counteracts Nesprin-2G-mediated mechanical forces during nuclear envelope repair.
- Author
-
Wallis SS, Ventimiglia LN, Otigbah E, Infante E, Cuesta-Geijo MA, Kidiyoor GR, Carbajal MA, Fleck RA, Foiani M, Garcia-Manyes S, Martin-Serrano J, and Agromayor M
- Subjects
- Actins chemistry, Cell Movement, Endosomal Sorting Complexes Required for Transport genetics, HeLa Cells, Humans, Mechanical Phenomena, Microfilament Proteins genetics, Nerve Tissue Proteins genetics, Cell Nucleus physiology, Cytoskeleton chemistry, Endosomal Sorting Complexes Required for Transport metabolism, Microfilament Proteins metabolism, Nerve Tissue Proteins metabolism, Nuclear Envelope physiology
- Abstract
Transient nuclear envelope ruptures during interphase (NERDI) occur due to cytoskeletal compressive forces at sites of weakened lamina, and delayed NERDI repair results in genomic instability. Nuclear envelope (NE) sealing is completed by endosomal sorting complex required for transport (ESCRT) machinery. A key unanswered question is how local compressive forces are counteracted to allow efficient membrane resealing. Here, we identify the ESCRT-associated protein BROX as a crucial factor required to accelerate repair of the NE. Critically, BROX binds Nesprin-2G, a component of the linker of nucleoskeleton and cytoskeleton complex (LINC). This interaction promotes Nesprin-2G ubiquitination and facilitates the relaxation of mechanical stress imposed by compressive actin fibers at the rupture site. Thus, BROX rebalances excessive cytoskeletal forces in cells experiencing NE instability to promote effective NERDI repair. Our results demonstrate that BROX coordinates mechanoregulation with membrane remodeling to ensure the maintenance of nuclear-cytoplasmic compartmentalization and genomic stability., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
50. A rapid method to visualize human mitochondrial DNA replication through rotary shadowing and transmission electron microscopy.
- Author
-
Kosar M, Piccini D, Foiani M, and Giannattasio M
- Subjects
- Humans, DNA Replication, DNA, Mitochondrial ultrastructure, DNA, Single-Stranded ultrastructure, Microscopy, Electron, Transmission methods, Mitochondria genetics, Mitochondria ultrastructure
- Abstract
We report a rapid experimental procedure based on high-density in vivo psoralen inter-strand DNA cross-linking coupled to spreading of naked purified DNA, positive staining, low-angle rotary shadowing, and transmission electron microscopy (TEM) that allows quick visualization of the dynamic of heavy strand (HS) and light strand (LS) human mitochondrial DNA replication. Replication maps built on linearized mitochondrial genomes and optimized rotary shadowing conditions enable clear visualization of the progression of the mitochondrial DNA synthesis and visualization of replication intermediates carrying long single-strand DNA stretches. One variant of this technique, called denaturing spreading, allowed the inspection of the fine chromatin structure of the mitochondrial genome and was applied to visualize the in vivo three-strand DNA structure of the human mitochondrial D-loop intermediate with unprecedented clarity., (© The Author(s) 2021. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.