63 results on '"Noberini R"'
Search Results
2. Advances in enrichment methods for mass spectrometry-based proteomics analysis of post-translational modifications
- Author
-
Brandi, J., Noberini, R., Bonaldi, T., and Cecconi, D.
- Subjects
Proteomics ,Proteome ,Chemical enrichment strategies ,Enrichment ,Immunoaffinity ,Mass spectrometry ,Post-translational modifications ,Organic Chemistry ,Acetylation ,General Medicine ,Biochemistry ,Mass Spectrometry ,Analytical Chemistry ,Settore BIO/13 - Biologia Applicata ,Settore BIO/10 - Biochimica ,Protein Processing, Post-Translational - Abstract
Post-translational modifications (PTMs) occur during or after protein biosynthesis and increase the functional diversity of proteome. They comprise phosphorylation, acetylation, methylation, glycosylation, ubiquitination, sumoylation (among many other modifications), and influence all aspects of cell biology. Mass-spectrometry (MS)-based proteomics is the most powerful approach for PTM analysis. Despite this, it is challenging due to low abundance and labile nature of many PTMs. Hence, enrichment of modified peptides is required for MS analysis. This review provides an overview of most common PTMs and a discussion of current enrichment methods for MS-based proteomics analysis. The traditional affinity strategies, including immunoenrichment, chromatography and protein pull-down, are outlined together with their strengths and shortcomings. Moreover, a special attention is paid to chemical enrichment strategies, such as capture by chemoselective probes, metabolic and chemoenzymatic labelling, which are discussed with an emphasis on their recent progress. Finally, the challenges and future trends in the field are discussed.
- Published
- 2022
3. Crystal structure of Eph A4 receptor
- Author
-
Qin, H., primary, Shi, J., additional, Noberini, R., additional, Pasquale, E.B., additional, and Song, J., additional
- Published
- 2008
- Full Text
- View/download PDF
4. Profiling of epigenetic features in clinical samples reveals novel widespread changes in cancer
- Author
-
Tiziana Bonaldi, Daniela Osti, Michela Lupia, Mohssen Ansarin, Francesco Raimondi, Giovanni Bertalot, Maria Giovanna Jodice, Evelyn Oliva Savoia, Maria Capra, Susanna Chiocca, Lavinia Ghiani, Marta Tagliabue, Camilla Restellini, Fausto Maffini, Giuseppina Bonizzi, Marco Giordano, Giuliana Pelicci, Roberta Noberini, Noberini, R., Restellini, C., Savoia, E. O., Raimondi, F., Ghiani, L., Jodice, M. G., Bertalot, G., Bonizzi, G., Capra, M., Maffini, F. A., Tagliabue, M., Ansarin, M., Lupia, M., Giordano, M., Osti, D., Pelicci, G., Chiocca, S., and Bonaldi, T.
- Subjects
0301 basic medicine ,Proteomics ,Cancer Research ,Histone-modifying enzymes ,Cell cycle ,lcsh:RC254-282 ,03 medical and health sciences ,Histone H3 ,0302 clinical medicine ,proteomics ,Gene expression ,cancer ,Epigenetics ,Histone post-translational modification ,mass spectrometry ,Cancer ,cell culture ,biology ,epigenetics ,Mass spectrometry ,Communication ,Epigenetic ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,Histone ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,histone post-translational modifications ,cell cycle - Abstract
Aberrations in histone post-translational modifications (PTMs), as well as in the histone modifying enzymes (HMEs) that catalyze their deposition and removal, have been reported in many tumors and many epigenetic inhibitors are currently under investigation for cancer treatment. Therefore, profiling epigenetic features in cancer could have important implications for the discovery of both biomarkers for patient stratification and novel epigenetic targets. In this study, we employed mass spectrometry-based approaches to comprehensively profile histone H3 PTMs in a panel of normal and tumoral tissues for different cancer types, identifying various changes, some of which appear to be a consequence of the increased proliferation rate of tumors, while others are cell-cycle independent. Histone PTM changes found in tumors partially correlate with alterations of the gene expression profiles of HMEs obtained from publicly available data and are generally lost in culture conditions. Through this analysis, we identified tumor- and subtype-specific histone PTM changes, but also widespread changes in the levels of histone H3 K9me3 and K14ac marks. In particular, H3K14ac showed a cell-cycle independent decrease in all the seven tumor/tumor subtype models tested and could represent a novel epigenetic hallmark of cancer.
- Published
- 2019
5. Age-associated metabolic and epigenetic barriers during direct reprogramming of mouse fibroblasts into induced cardiomyocytes.
- Author
-
Santos F, Correia M, Dias R, Bola B, Noberini R, Ferreira RS, Trigo D, Domingues P, Teixeira J, Bonaldi T, Oliveira PJ, Bär C, de Jesus BB, and Nóbrega-Pereira S
- Abstract
Heart disease is the leading cause of mortality in developed countries, and novel regenerative procedures are warranted. Direct cardiac conversion (DCC) of adult fibroblasts can create induced cardiomyocytes (iCMs) for gene and cell-based heart therapy, and in addition to holding great promise, still lacks effectiveness as metabolic and age-associated barriers remain elusive. Here, by employing MGT (Mef2c, Gata4, Tbx5) transduction of mouse embryonic fibroblasts (MEFs) and adult (dermal and cardiac) fibroblasts from animals of different ages, we provide evidence that the direct reprogramming of fibroblasts into iCMs decreases with age. Analyses of histone posttranslational modifications and ChIP-qPCR revealed age-dependent alterations in the epigenetic landscape of DCC. Moreover, DCC is accompanied by profound mitochondrial metabolic adaptations, including a lower abundance of anabolic metabolites, network remodeling, and reliance on mitochondrial respiration. In vitro metabolic modulation and dietary manipulation in vivo improve DCC efficiency and are accompanied by significant alterations in histone marks and mitochondrial homeostasis. Importantly, adult-derived iCMs exhibit increased accumulation of oxidative stress in the mitochondria and activation of mitophagy or dietary lipids; they improve DCC and revert mitochondrial oxidative damage. Our study provides evidence that metaboloepigenetics plays a direct role in cell fate transitions driving DCC, highlighting the potential use of metabolic modulation to improve cardiac regenerative strategies., (© 2024 The Author(s). Aging Cell published by Anatomical Society and John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
6. Multiscale modeling uncovers 7q11.23 copy number variation-dependent changes in ribosomal biogenesis and neuronal maturation and excitability.
- Author
-
Mihailovich M, Germain PL, Shyti R, Pozzi D, Noberini R, Liu Y, Aprile D, Tenderini E, Troglio F, Trattaro S, Fabris S, Ciptasari U, Rigoli MT, Caporale N, D'Agostino G, Mirabella F, Vitriolo A, Capocefalo D, Skaros A, Franchini AV, Ricciardi S, Biunno I, Neri A, Nadif Kasri N, Bonaldi T, Aebersold R, Matteoli M, and Testa G
- Subjects
- Humans, Ribosomes metabolism, Ribosomes genetics, Neurogenesis genetics, Williams Syndrome genetics, Williams Syndrome metabolism, Williams Syndrome pathology, Williams Syndrome physiopathology, Ribosomal Protein S6 metabolism, Ribosomal Protein S6 genetics, TOR Serine-Threonine Kinases metabolism, TOR Serine-Threonine Kinases genetics, Male, Cell Differentiation, Female, DNA Copy Number Variations, Neurons metabolism, Neurons pathology, Chromosomes, Human, Pair 7 genetics
- Abstract
Copy number variation (CNV) at 7q11.23 causes Williams-Beuren syndrome (WBS) and 7q microduplication syndrome (7Dup), neurodevelopmental disorders (NDDs) featuring intellectual disability accompanied by symmetrically opposite neurocognitive features. Although significant progress has been made in understanding the molecular mechanisms underlying 7q11.23-related pathophysiology, the propagation of CNV dosage across gene expression layers and their interplay remains elusive. Here we uncovered 7q11.23 dosage-dependent symmetrically opposite dynamics in neuronal differentiation and intrinsic excitability. By integrating transcriptomics, translatomics, and proteomics of patient-derived and isogenic induced neurons, we found that genes related to neuronal transmission follow 7q11.23 dosage and are transcriptionally controlled, while translational factors and ribosomal genes are posttranscriptionally buffered. Consistently, we found phosphorylated RPS6 (p-RPS6) downregulated in WBS and upregulated in 7Dup. Surprisingly, p-4EBP was changed in the opposite direction, reflecting dosage-specific changes in total 4EBP levels. This highlights different dosage-sensitive dyregulations of the mTOR pathway as well as distinct roles of p-RPS6 and p-4EBP during neurogenesis. Our work demonstrates the importance of multiscale disease modeling across molecular and functional layers, uncovers the pathophysiological relevance of ribosomal biogenesis in a paradigmatic pair of NDDs, and uncouples the roles of p-RPS6 and p-4EBP as mechanistically actionable relays in NDDs.
- Published
- 2024
- Full Text
- View/download PDF
7. Improved Mass Spectrometry-Based Methods Reveal Abundant Propionylation and Tissue-Specific Histone Propionylation Profiles.
- Author
-
Vai A, Noberini R, Ghirardi C, Rodrigues de Paula D, Carminati M, Pallavi R, Araújo N, Varga-Weisz P, and Bonaldi T
- Subjects
- Humans, Animals, Mice, Female, Mass Spectrometry methods, Acylation, Organ Specificity, Acetylation, Proteomics methods, Histones metabolism, Protein Processing, Post-Translational, Breast Neoplasms metabolism
- Abstract
Histone posttranslational modifications (PTMs) have crucial roles in a multitude of cellular processes, and their aberrant levels have been linked with numerous diseases, including cancer. Although histone PTM investigations have focused so far on methylations and acetylations, alternative long-chain acylations emerged as new dimension, as they are linked to cellular metabolic states and affect gene expression through mechanisms distinct from those regulated by acetylation. Mass spectrometry is the most powerful, comprehensive, and unbiased method to study histone PTMs. However, typical mass spectrometry-based protocols for histone PTM analysis do not allow the identification of naturally occurring propionylation and butyrylation. Here, we present improved state-of-the-art sample preparation and analysis protocols to quantitate these classes of modifications. After testing different derivatization methods coupled to protease digestion, we profiled common histone PTMs and histone acylations in seven mouse tissues and human normal and tumor breast clinical samples, obtaining a map of propionylations and butyrylations found in different tissue contexts. A quantitative histone PTM analysis also revealed a contribution of histone acylations in discriminating different tissues, also upon perturbation with antibiotics, and breast cancer samples from the normal counterpart. Our results show that profiling only classical modifications is limiting and highlight the importance of using sample preparation methods that allow the analysis of the widest possible spectrum of histone modifications, paving the way for deeper insights into their functional significance in cellular processes and disease states., Competing Interests: Conflicts of interest A. V. and M. C. are PhD students within the European School of Molecular Medicine (SEMM). The other authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. Long-term Multimodal Recording Reveals Epigenetic Adaptation Routes in Dormant Breast Cancer Cells.
- Author
-
Rosano D, Sofyali E, Dhiman H, Ghirardi C, Ivanoiu D, Heide T, Vingiani A, Bertolotti A, Pruneri G, Canale E, Dewhurst HF, Saha D, Slaven N, Barozzi I, Li T, Zemlyanskiy G, Phillips H, James C, Győrffy B, Lynn C, Cresswell GD, Rehman F, Noberini R, Bonaldi T, Sottoriva A, and Magnani L
- Subjects
- Humans, Female, Neoplasm Recurrence, Local genetics, Gene Expression Regulation, Neoplastic, Breast Neoplasms genetics, Breast Neoplasms pathology, Breast Neoplasms drug therapy, Epigenesis, Genetic
- Abstract
Patients with estrogen receptor-positive breast cancer receive adjuvant endocrine therapies (ET) that delay relapse by targeting clinically undetectable micrometastatic deposits. Yet, up to 50% of patients relapse even decades after surgery through unknown mechanisms likely involving dormancy. To investigate genetic and transcriptional changes underlying tumor awakening, we analyzed late relapse patients and longitudinally profiled a rare cohort treated with long-term neoadjuvant ETs until progression. Next, we developed an in vitro evolutionary study to record the adaptive strategies of individual lineages in unperturbed parallel experiments. Our data demonstrate that ETs induce nongenetic cell state transitions into dormancy in a stochastic subset of cells via epigenetic reprogramming. Single lineages with divergent phenotypes awaken unpredictably in the absence of recurrent genetic alterations. Targeting the dormant epigenome shows promising activity against adapting cancer cells. Overall, this study uncovers the contribution of epigenetic adaptation to the evolution of resistance to ETs., Significance: This study advances the understanding of therapy-induced dormancy with potential clinical implications for breast cancer. Estrogen receptor-positive breast cancer cells adapt to endocrine treatment by entering a dormant state characterized by strong heterochromatinization with no recurrent genetic changes. Targeting the epigenetic rewiring impairs the adaptation of cancer cells to ETs. See related commentary by Llinas-Bertran et al., p. 704. This article is featured in Selected Articles from This Issue, p. 695., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
9. Hyperacetylated histone H4 is a source of carbon contributing to lipid synthesis.
- Author
-
Charidemou E, Noberini R, Ghirardi C, Georgiou P, Marcou P, Theophanous A, Strati K, Keun H, Behrends V, Bonaldi T, and Kirmizis A
- Subjects
- Animals, Mice, Lipogenesis, Chromatin, Acetyltransferases metabolism, Lipids, Acetylation, Histone Acetyltransferases genetics, Histone Acetyltransferases metabolism, Histones metabolism, Carbon metabolism
- Abstract
Histone modifications commonly integrate environmental cues with cellular metabolic outputs by affecting gene expression. However, chromatin modifications such as acetylation do not always correlate with transcription, pointing towards an alternative role of histone modifications in cellular metabolism. Using an approach that integrates mass spectrometry-based histone modification mapping and metabolomics with stable isotope tracers, we demonstrate that elevated lipids in acetyltransferase-depleted hepatocytes result from carbon atoms derived from deacetylation of hyperacetylated histone H4 flowing towards fatty acids. Consistently, enhanced lipid synthesis in acetyltransferase-depleted hepatocytes is dependent on histone deacetylases and acetyl-CoA synthetase ACSS2, but not on the substrate specificity of the acetyltransferases. Furthermore, we show that during diet-induced lipid synthesis the levels of hyperacetylated histone H4 decrease in hepatocytes and in mouse liver. In addition, overexpression of acetyltransferases can reverse diet-induced lipogenesis by blocking lipid droplet accumulation and maintaining the levels of hyperacetylated histone H4. Overall, these findings highlight hyperacetylated histones as a metabolite reservoir that can directly contribute carbon to lipid synthesis, constituting a novel function of chromatin in cellular metabolism., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
10. Activation of endogenous retroviruses and induction of viral mimicry by MEK1/2 inhibition in pancreatic cancer.
- Author
-
Cortesi A, Gandolfi F, Arco F, Di Chiaro P, Valli E, Polletti S, Noberini R, Gualdrini F, Attanasio S, Citron F, Ho IL, Shah R, Yen EY, Spinella MC, Ronzoni S, Rodighiero S, Mitro N, Bonaldi T, Ghisletti S, Monticelli S, Viale A, Diaferia GR, and Natoli G
- Subjects
- Humans, Signal Transduction, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Endogenous Retroviruses genetics, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism
- Abstract
While pancreatic ductal adenocarcinomas (PDACs) are addicted to KRAS-activating mutations, inhibitors of downstream KRAS effectors, such as the MEK1/2 kinase inhibitor trametinib, are devoid of therapeutic effects. However, the extensive rewiring of regulatory circuits driven by the attenuation of the KRAS pathway may induce vulnerabilities of therapeutic relevance. An in-depth molecular analysis of the transcriptional and epigenomic alterations occurring in PDAC cells in the initial hours after MEK1/2 inhibition by trametinib unveiled the induction of endogenous retroviruses (ERVs) escaping epigenetic silencing, leading to the production of double-stranded RNAs and the increased expression of interferon (IFN) genes. We tracked ERV activation to the early induction of the transcription factor ELF3, which extensively bound and activated nonsilenced retroelements and synergized with IRF1 (interferon regulatory factor 1) in the activation of IFNs and IFN-stimulated genes. Trametinib-induced viral mimicry in PDAC may be exploited in the rational design of combination therapies in immuno-oncology.
- Published
- 2024
- Full Text
- View/download PDF
11. Acetyl-CoA production by Mediator-bound 2-ketoacid dehydrogenases boosts de novo histone acetylation and is regulated by nitric oxide.
- Author
-
Russo M, Gualdrini F, Vallelonga V, Prosperini E, Noberini R, Pedretti S, Borriero C, Di Chiaro P, Polletti S, Imperato G, Marenda M, Ghirardi C, Bedin F, Cuomo A, Rodighiero S, Bonaldi T, Mitro N, Ghisletti S, and Natoli G
- Subjects
- Acetyl Coenzyme A metabolism, Acetylation, Mediator Complex metabolism, Oxidoreductases metabolism, Histones genetics, Histones metabolism, Nitric Oxide metabolism
- Abstract
Histone-modifying enzymes depend on the availability of cofactors, with acetyl-coenzyme A (CoA) being required for histone acetyltransferase (HAT) activity. The discovery that mitochondrial acyl-CoA-producing enzymes translocate to the nucleus suggests that high concentrations of locally synthesized metabolites may impact acylation of histones and other nuclear substrates, thereby controlling gene expression. Here, we show that 2-ketoacid dehydrogenases are stably associated with the Mediator complex, thus providing a local supply of acetyl-CoA and increasing the generation of hyper-acetylated histone tails. Nitric oxide (NO), which is produced in large amounts in lipopolysaccharide-stimulated macrophages, inhibited the activity of Mediator-associated 2-ketoacid dehydrogenases. Elevation of NO levels and the disruption of Mediator complex integrity both affected de novo histone acetylation within a shared set of genomic regions. Our findings indicate that the local supply of acetyl-CoA generated by 2-ketoacid dehydrogenases bound to Mediator is required to maximize acetylation of histone tails at sites of elevated HAT activity., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
12. Mass Spectrometry-Based Profiling of Histone Post-Translational Modifications in Uveal Melanoma Tissues, Human Melanocytes, and Uveal Melanoma Cell Lines - A Pilot Study.
- Author
-
Herwig-Carl MC, Sharma A, Tischler V, Pelusi N, Loeffler KU, Holz FG, Zeschnigk M, Landreville S, Auw-Haedrich C, Noberini R, and Bonaldi T
- Subjects
- Humans, Histones, Pilot Projects, Melanocytes metabolism, Cell Line, Mass Spectrometry, Melanoma metabolism, Uveal Neoplasms pathology
- Abstract
Purpose: Epigenetic alterations in uveal melanoma (UM) are still neither well characterized, nor understood. In this pilot study, we sought to provide a deeper insight into the possible role of epigenetic alterations in the pathogenesis of UM and their potential prognostic relevance. To this aim, we comprehensively profiled histone post-translational modifications (PTMs), which represent epigenetic features regulating chromatin accessibility and gene transcription, in UM formalin-fixed paraffin-embedded (FFPE) tissues, control tissues, UM cell lines, and healthy melanocytes., Methods: FFPE tissues of UM (n = 24), normal choroid (n = 4), human UM cell lines (n = 7), skin melanocytes (n = 6), and uveal melanocytes (n = 2) were analyzed through a quantitative liquid chromatography-mass spectrometry (LC-MS) approach., Results: Hierarchical clustering showed a clear separation with several histone PTMs that changed significantly in a tumor compared to normal samples, in both tissues and cell lines. In addition, several acetylations and H4K20me1 showed lower levels in BAP1 mutant tumors. Some of these changes were also observed when we compared GNA11 mutant tumors with GNAQ tumors. The epigenetic profiling of cell lines revealed that the UM cell lines MP65 and UPMM1 have a histone PTM pattern closer to the primary tissues than the other cell lines analyzed., Conclusions: Our results suggest the existence of different histone PTM patterns that may be important for diagnosis and prognosis in UM. However, further analyses are needed to confirm these findings in a larger cohort. The epigenetic characterization of a panel of UM cell lines suggested which cellular models are more suitable for epigenetic investigations.
- Published
- 2024
- Full Text
- View/download PDF
13. A comprehensive characterisation of phaeochromocytoma and paraganglioma tumours through histone protein profiling, DNA methylation and transcriptomic analysis genome wide.
- Author
-
Chatzikyriakou P, Brempou D, Quinn M, Fishbein L, Noberini R, Anastopoulos IN, Tufton N, Lim ES, Obholzer R, Hubbard JG, Moonim M, Bonaldi T, Nathanson KL, Izatt L, and Oakey RJ
- Subjects
- Humans, Histones genetics, Histones metabolism, DNA Methylation, Gene Expression Profiling, Pheochromocytoma genetics, Pheochromocytoma metabolism, Pheochromocytoma pathology, Paraganglioma genetics, Paraganglioma pathology, Adrenal Gland Neoplasms genetics, Adrenal Gland Neoplasms metabolism, Adrenal Gland Neoplasms pathology
- Abstract
Background: Phaeochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine tumours. Pathogenic variants have been identified in more than 15 susceptibility genes; associated tumours are grouped into three Clusters, reinforced by their transcriptional profiles. Cluster 1A PPGLs have pathogenic variants affecting enzymes of the tricarboxylic acid cycle, including succinate dehydrogenase. Within inherited PPGLs, these are the most common. PPGL tumours are known to undergo epigenetic reprograming, and here, we report on global histone post-translational modifications and DNA methylation levels, alongside clinical phenotypes., Results: Out of the 25 histone post-translational modifications examined, Cluster 1A PPGLs were distinguished from other tumours by a decrease in hyper-acetylated peptides and an increase in H3K4me2. DNA methylation was compared between tumours from individuals who developed metastatic disease versus those that did not. The majority of differentially methylated sites identified tended to be completely methylated or unmethylated in non-metastatic tumours, with low inter-sample variance. Metastatic tumours by contrast consistently had an intermediate DNA methylation state, including the ephrin receptor EPHA4 and its ligand EFNA3. Gene expression analyses performed to identify genes involved in metastatic tumour behaviour pin-pointed a number of genes previously described as mis-regulated in Cluster 1A tumours, as well as highlighting the tumour suppressor RGS22 and the pituitary tumour-transforming gene PTTG1., Conclusions: Combined transcriptomic and DNA methylation analyses revealed aberrant pathways, including ones that could be implicated in metastatic phenotypes and, for the first time, we report a decrease in hyper-acetylated histone marks in Cluster 1 PPGLs., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
14. Multi-omics integrative modelling for stereotactic body radiotherapy in early-stage non-small cell lung cancer: clinical trial protocol of the MONDRIAN study.
- Author
-
Volpe S, Zaffaroni M, Piperno G, Vincini MG, Zerella MA, Mastroleo F, Cattani F, Fodor CI, Bellerba F, Bonaldi T, Bonizzi G, Ceci F, Cremonesi M, Fusco N, Gandini S, Garibaldi C, Torre D, Noberini R, Petralia G, Spaggiari L, Venetis K, Orecchia R, Casiraghi M, and Jereczek-Fossa BA
- Subjects
- Humans, Multiomics, Neoplasm Staging, Observational Studies as Topic, Proteomics, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung radiotherapy, Carcinoma, Non-Small-Cell Lung pathology, Lung Neoplasms genetics, Lung Neoplasms radiotherapy, Lung Neoplasms pathology, Radiosurgery methods, Small Cell Lung Carcinoma
- Abstract
Background: Currently, main treatment strategies for early-stage non-small cell lung cancer (ES-NSCLC) disease are surgery or stereotactic body radiation therapy (SBRT), with successful local control rates for both approaches. However, regional and distant failure remain critical in SBRT, and it is paramount to identify predictive factors of response to identify high-risk patients who may benefit from more aggressive approaches. The main endpoint of the MONDRIAN trial is to identify multi-omic biomarkers of SBRT response integrating information from the individual fields of radiomics, genomics and proteomics., Methods: MONDRIAN is a prospective observational explorative cohort clinical study, with a data-driven, bottom-up approach. It is expected to enroll 100 ES-NSCLC SBRT candidates treated at an Italian tertiary cancer center with well-recognized expertise in SBRT and thoracic surgery. To identify predictors specific to SBRT, MONDRIAN will include data from 200 patients treated with surgery, in a 1:2 ratio, with comparable clinical characteristics. The project will have an overall expected duration of 60 months, and will be structured into five main tasks: (i) Clinical Study; (ii) Imaging/ Radiomic Study, (iii) Gene Expression Study, (iv) Proteomic Study, (v) Integrative Model Building., Discussion: Thanks to its multi-disciplinary nature, MONDRIAN is expected to provide the opportunity to characterize ES-NSCLC from a multi-omic perspective, with a Radiation Oncology-oriented focus. Other than contributing to a mechanistic understanding of the disease, the study will assist the identification of high-risk patients in a largely unexplored clinical setting. Ultimately, this would orient further clinical research efforts on the combination of SBRT and systemic treatments, such as immunotherapy, with the perspective of improving oncological outcomes in this subset of patients., Trial Registration: The study was prospectively registered at clinicaltrials.gov (NCT05974475)., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
15. Proteomics contributions to epigenetic drug discovery.
- Author
-
Noberini R and Bonaldi T
- Subjects
- DNA Methylation, Epigenesis, Genetic, Protein Processing, Post-Translational, Histones metabolism, Proteomics methods
- Abstract
The combined activity of epigenetic features, which include histone post-translational modifications, DNA methylation, and nucleosome positioning, regulates gene expression independently from changes in the DNA sequence, defining how the shared genetic information of an organism is used to generate different cell phenotypes. Alterations in epigenetic processes have been linked with a multitude of diseases, including cancer, fueling interest in the discovery of drugs targeting the proteins responsible for writing, erasing, or reading histone and DNA modifications. Mass spectrometry (MS)-based proteomics has emerged as a versatile tool that can assist drug discovery pipelines from target validation, through target deconvolution, to monitoring drug efficacy in vivo. Here, we provide an overview of the contributions of MS-based proteomics to epigenetic drug discovery, describing the main approaches that can be used to support different drug discovery pipelines and highlighting how they contributed to the development and characterization of epigenetic drugs., (© 2023 The Authors. Proteomics published by Wiley-VCH GmbH.)
- Published
- 2023
- Full Text
- View/download PDF
16. A truncated and catalytically inactive isoform of KDM5B histone demethylase accumulates in breast cancer cells and regulates H3K4 tri-methylation and gene expression.
- Author
-
Di Nisio E, Licursi V, Mannironi C, Buglioni V, Paiardini A, Robusti G, Noberini R, Bonaldi T, and Negri R
- Subjects
- Humans, Female, Methylation, Histone Demethylases genetics, Histone Demethylases metabolism, Protein Isoforms genetics, Protein Isoforms metabolism, Gene Expression, Nuclear Proteins metabolism, Repressor Proteins metabolism, Jumonji Domain-Containing Histone Demethylases genetics, Jumonji Domain-Containing Histone Demethylases metabolism, Histones genetics, Breast Neoplasms genetics
- Abstract
KDM5B histone demethylase is overexpressed in many cancers and plays an ambivalent role in oncogenesis, depending on the specific context. This ambivalence could be explained by the expression of KDM5B protein isoforms with diverse functional roles, which could be present at different levels in various cancer cell lines. We show here that one of these isoforms, namely KDM5B-NTT, accumulates in breast cancer cell lines due to remarkable protein stability relative to the canonical PLU-1 isoform, which shows a much faster turnover. This isoform is the truncated and catalytically inactive product of an mRNA with a transcription start site downstream of the PLU-1 isoform, and the consequent usage of an alternative ATG for translation initiation. It also differs from the PLU-1 transcript in the inclusion of an additional exon (exon-6), previously attributed to other putative isoforms. Overexpression of this isoform in MCF7 cells leads to an increase in bulk H3K4 methylation and induces derepression of a gene cluster, including the tumor suppressor Cav1 and several genes involved in the interferon-alpha and -gamma response. We discuss the relevance of this finding considering the hypothesis that KDM5B may possess regulatory roles independent of its catalytic activity., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
17. Detection and quantification of the histone code in the fungal genus Aspergillus.
- Author
-
Zhang X, Noberini R, Vai A, Bonaldi T, Seidl MF, and Collemare J
- Subjects
- Histone Code genetics, Protein Processing, Post-Translational, Heterochromatin, Histones genetics, Histones metabolism, Aspergillus nidulans genetics, Aspergillus nidulans metabolism
- Abstract
In eukaryotes, the combination of different histone post-translational modifications (PTMs) - the histone code - impacts the chromatin organization as compact and transcriptionally silent heterochromatin or accessible and transcriptionally active euchromatin. Although specific histone PTMs have been studied in fungi, an overview of histone PTMs and their relative abundance is still lacking. Here, we used mass spectrometry to detect and quantify histone PTMs in three fungal species belonging to three distinct taxonomic sections of the genus Aspergillus (Aspergillus niger, Aspergillus nidulans (two strains), and Aspergillus fumigatus). We overall detected 23 different histone PTMs, including a majority of lysine methylations and acetylations, and 23 co-occurrence patterns of multiple histone PTMs. Among those, we report for the first time the detection of H3K79me1, H3K79me2, and H4K31ac in Aspergilli. Although all three species harbour the same PTMs, we found significant differences in the relative abundance of H3K9me1/2/3, H3K14ac, H3K36me1 and H3K79me1, as well as the co-occurrence of acetylation on both K18 and K23 of histone H3 in a strain-specific manner. Our results provide novel insights about the underexplored complexity of the histone code in filamentous fungi, and its functional implications on genome architecture and gene regulation., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
18. Small molecule-induced epigenomic reprogramming of APL blasts leading to antiviral-like response and c-MYC downregulation.
- Author
-
Amatori S, Persico G, Cantatore F, Rusin M, Formica M, Giorgi L, Macedi E, Casciaro F, Errico Provenzano A, Gambardella S, Noberini R, Bonaldi T, Fusi V, Giorgio M, and Fanelli M
- Subjects
- Humans, Histones genetics, Down-Regulation, Antiviral Agents pharmacology, Epigenomics, Lysine genetics, Lysine metabolism, Lysine pharmacology, Oncogene Proteins, Fusion genetics, Cell Differentiation, Leukemia, Promyelocytic, Acute genetics, Leukemia, Promyelocytic, Acute metabolism, Leukemia, Myeloid, Acute genetics
- Abstract
Acute promyelocytic leukemia (APL) is an aggressive subtype of acute myeloid leukemia (AML) in which the PML/RARα fusion protein exerts oncogenic activities by recruiting repressive complexes to the promoter of specific target genes. Other epigenetic perturbations, as alterations of histone H3 lysine 9 trimethylation (H3K9me3), have been frequently found in AMLs and are associated with leukemogenesis and leukemia progression. Here, we characterized the epigenomic effects of maltonis, a novel maltol-derived molecule, in APL cells. We demonstrate that maltonis treatments induce a profound remodulation of the histone code, reducing global H3K9me3 signal and modulating other histone post-translational modifications. Transcriptomic and epigenomic analyses revealed that maltonis exposure induces changes of genes expression associated with a genomic redistribution of histone H3 lysine 4 trimethylation (H3K4me3) and lysine 27 acetylation (H3K27ac). Upregulation of interferon alpha and gamma response and downregulation of c-MYC target genes, in function of c-MYC reduced expression (monitored in all the hematopoietic neoplasms tested), represent the most significant modulated pathways. These data demonstrate the ability of maltonis to epigenetically reprogram the gene expression profile of APL cells, inducing an intriguing antiviral-like response, concomitantly with the downregulation of c-MYC-related pathways, thus making it an attractive candidate for antileukemic therapy., (© 2022. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
19. Serum proteomics profiling identifies a preliminary signature for the diagnosis of early-stage lung cancer.
- Author
-
Gasparri R, Noberini R, Cuomo A, Yadav A, Tricarico D, Salvetto C, Maisonneuve P, Caminiti V, Sedda G, Sabalic A, Bonaldi T, and Spaggiari L
- Subjects
- Humans, Biomarkers, Tumor metabolism, Lung metabolism, Mass Spectrometry, Proteomics methods, Lung Neoplasms diagnosis, Lung Neoplasms genetics, Lung Neoplasms metabolism
- Abstract
Purpose: Lung cancer is the most common cause of death from cancer worldwide, largely due to late diagnosis. Thus, there is an urgent need to develop new approaches to improve the detection of early-stage lung cancer, which would greatly improve patient survival., Experimental Design: The quantitative protein expression profiles of microvesicles isolated from the sera from 46 lung cancer patients and 41 high-risk non-cancer subjects were obtained using a mass spectrometry method based on a peptide library matching approach., Results: We identified 33 differentially expressed proteins that allow discriminating the two groups. We also built a machine learning model based on serum protein expression profiles that can correctly classify the majority of lung cancer cases and that highlighted a decrease in the levels of Arysulfatase A (ARSA) as the most discriminating factor found in tumors., Conclusions and Clinical Relevance: Our study identified a preliminary, non-invasive protein signature able to discriminate with high specificity and selectivity early-stage lung cancer patients from high-risk healthy subjects. These results provide the basis for future validation studies for the development of a non-invasive diagnostic tool for lung cancer., (© 2023 The Authors. Proteomics - Clinical Applications published by Wiley-VCH GmbH.)
- Published
- 2023
- Full Text
- View/download PDF
20. A Super-SILAC Approach for Profiling Histone Posttranslational Modifications.
- Author
-
Noberini R, Longhi E, and Bonaldi T
- Subjects
- Isotope Labeling methods, Protein Processing, Post-Translational, Mass Spectrometry methods, Histones metabolism, Proteomics methods
- Abstract
Histone posttranslational modifications (PTMs) play an important role in the regulation of gene expression and have been implicated in a multitude of physiological and pathological processes. During the last decade, mass spectrometry (MS) has emerged as the most accurate and versatile tool to quantitate histone PTMs. Stable-isotope labeling by amino acids in cell culture (SILAC) is an MS-based quantitation strategy involving metabolic labeling of cells, which has been applied to global protein profiling as well as histone PTM analysis. The classical SILAC approach is associated with reduced experimental variability and high quantitation accuracy, but provides limited multiplexing capabilities and can be applied only to actively dividing cells, thus excluding clinical samples. Both limitations are overcome by an evolution of classical SILAC involving the use of a mix of heavy-labeled cell lines as a spike-in standard, known as "super-SILAC". In this chapter, we will provide a detailed description of the optimized protocol used in our laboratory to generate a histone-focused super-SILAC mix and employ it as an internal standard for histone PTM quantitation., (© 2023. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
21. Mass Spectrometry-Based Analysis of Histone Posttranslational Modifications from Laser Microdissected Samples.
- Author
-
Noberini R and Bonaldi T
- Subjects
- Protein Processing, Post-Translational, Lasers, Mass Spectrometry, Histones, Histone Code
- Abstract
The analysis of histone posttranslational modifications (PTMs) in clinical samples has gained considerable interest due to the increasing knowledge about the implication of epigenetics in a multitude of physiological and pathological processes. Mass spectrometry (MS) has emerged as the most accurate and versatile tool to detect and quantify histone PTMs and has also been applied to clinical specimens, thanks to protocols developed during the past years. However, the requirement for relatively large amounts of material has so far impaired the application of these approaches to samples available in limited amounts. To address this issue, we have recently streamlined the protein extraction procedure from low-amount clinical samples and optimized the digestion step, obtaining a protocol suitable for the analysis of the most common histone PTMs from laser microdissected tissue areas containing down to 1000 cells, which we will describe in this chapter., (© 2023. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
22. Investigating pathological epigenetic aberrations by epi-proteomics.
- Author
-
Robusti G, Vai A, Bonaldi T, and Noberini R
- Subjects
- Humans, DNA Methylation, Epigenomics, Protein Processing, Post-Translational, Epigenesis, Genetic, Histones metabolism, Proteomics methods
- Abstract
Epigenetics includes a complex set of processes that alter gene activity without modifying the DNA sequence, which ultimately determines how the genetic information common to all the cells of an organism is used to generate different cell types. Dysregulation in the deposition and maintenance of epigenetic features, which include histone posttranslational modifications (PTMs) and histone variants, can result in the inappropriate expression or silencing of genes, often leading to diseased states, including cancer. The investigation of histone PTMs and variants in the context of clinical samples has highlighted their importance as biomarkers for patient stratification and as key players in aberrant epigenetic mechanisms potentially targetable for therapy. Mass spectrometry (MS) has emerged as the most powerful and versatile tool for the comprehensive, unbiased and quantitative analysis of histone proteoforms. In recent years, these approaches-which we refer to as "epi-proteomics"-have demonstrated their usefulness for the investigation of epigenetic mechanisms in pathological conditions, offering a number of advantages compared with the antibody-based methods traditionally used to profile clinical samples. In this review article, we will provide a critical overview of the MS-based approaches that can be employed to study histone PTMs and variants in clinical samples, with a strong focus on the latest advances in this area, such as the analysis of uncommon modifications and the integration of epi-proteomics data into multi-OMICs approaches, as well as the challenges to be addressed to fully exploit the potential of this novel field of research., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
23. The histone code of the fungal genus Aspergillus uncovered by evolutionary and proteomic analyses.
- Author
-
Zhang X, Noberini R, Bonaldi T, Collemare J, and Seidl MF
- Subjects
- Chromatin, DNA, Histones genetics, Histones metabolism, Lysine genetics, Lysine metabolism, Phylogeny, Polycomb Repressive Complex 2 genetics, Polycomb Repressive Complex 2 metabolism, Proteomics, Aspergillus nidulans genetics, Aspergillus nidulans metabolism, Histone Code genetics
- Abstract
Chemical modifications of DNA and histone proteins impact the organization of chromatin within the nucleus. Changes in these modifications, catalysed by different chromatin-modifying enzymes, influence chromatin organization, which in turn is thought to impact the spatial and temporal regulation of gene expression. While combinations of different histone modifications, the histone code, have been studied in several model species, we know very little about histone modifications in the fungal genus Aspergillus , whose members are generally well studied due to their importance as models in cell and molecular biology as well as their medical and biotechnological relevance. Here, we used phylogenetic analyses in 94 Aspergilli as well as other fungi to uncover the occurrence and evolutionary trajectories of enzymes and protein complexes with roles in chromatin modifications or regulation. We found that these enzymes and complexes are highly conserved in Aspergilli, pointing towards a complex repertoire of chromatin modifications. Nevertheless, we also observed few recent gene duplications or losses, highlighting Aspergillus species to further study the roles of specific chromatin modifications. SET7 (KMT6) and other components of PRC2 (Polycomb Repressive Complex 2), which is responsible for methylation on histone H3 at lysine 27 in many eukaryotes including fungi, are absent in Aspergilli as well as in closely related Penicillium species, suggesting that these lost the capacity for this histone modification. We corroborated our computational predictions by performing untargeted MS analysis of histone post-translational modifications in Aspergillus nidulans . This systematic analysis will pave the way for future research into the complexity of the histone code and its functional implications on genome architecture and gene regulation in fungi.
- Published
- 2022
- Full Text
- View/download PDF
24. Mass spectrometry-based characterization of histones in clinical samples: applications, progress, and challenges.
- Author
-
Noberini R, Robusti G, and Bonaldi T
- Subjects
- Antineoplastic Agents therapeutic use, Chromatin chemistry, Chromatin metabolism, Gene Expression Regulation, Neoplastic, Histones metabolism, Humans, Mass Spectrometry methods, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Neoplasms diagnosis, Neoplasms drug therapy, Neoplasms pathology, Peptide Mapping methods, Peptides genetics, Peptides metabolism, Proteomics, Epigenesis, Genetic, Histones genetics, Mass Spectrometry trends, Neoplasms genetics, Peptides analysis, Protein Processing, Post-Translational
- Abstract
In the last 15 years, increasing evidence linking epigenetics to various aspects of cancer biology has prompted the investigation of histone post-translational modifications (PTMs) and histone variants in the context of clinical samples. The studies performed so far demonstrated the potential of this type of investigations for the discovery of both potential epigenetic biomarkers for patient stratification and novel epigenetic mechanisms potentially targetable for cancer therapy. Although traditionally the analysis of histones in clinical samples was performed through antibody-based methods, mass spectrometry (MS) has emerged as a more powerful tool for the unbiased, comprehensive, and quantitative investigation of histone PTMs and variants. MS has been extensively used for the analysis of epigenetic marks in cell lines and animal tissue and, thanks to recent technological advances, is now ready to be applied also to clinical samples. In this review, we will provide an overview on the quantitative MS-based analysis of histones, their PTMs and their variants in cancer clinical samples, highlighting current achievements and future perspectives for this novel field of research. Among the different MS-based approaches currently available for histone PTM profiling, we will focus on the 'bottom-up' strategy, namely the analysis of short proteolytic peptides, as it has been already successfully employed for the analysis of clinical samples., (© 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2022
- Full Text
- View/download PDF
25. LSD1-directed therapy affects glioblastoma tumorigenicity by deregulating the protective ATF4-dependent integrated stress response.
- Author
-
Faletti S, Osti D, Ceccacci E, Richichi C, Costanza B, Nicosia L, Noberini R, Marotta G, Furia L, Faretta MR, Brambillasca S, Quarto M, Bertero L, Boldorini R, Pollo B, Gandini S, Cora D, Minucci S, Mercurio C, Varasi M, Bonaldi T, and Pelicci G
- Subjects
- Activating Transcription Factor 4 metabolism, Cell Line, Tumor, Cell Proliferation, Histone Demethylases metabolism, Humans, Neoplasm Recurrence, Local metabolism, Neoplastic Stem Cells pathology, Brain Neoplasms drug therapy, Brain Neoplasms genetics, Brain Neoplasms metabolism, Glioblastoma drug therapy, Glioblastoma genetics, Glioblastoma metabolism
- Abstract
Glioblastoma (GBM) is a fatal tumor whose aggressiveness, heterogeneity, poor blood-brain barrier penetration, and resistance to therapy highlight the need for new targets and clinical treatments. A step toward clinical translation includes the eradication of GBM tumor-initiating cells (TICs), responsible for GBM heterogeneity and relapse. By using patient-derived TICs and xenograft orthotopic models, we demonstrated that the selective lysine-specific histone demethylase 1 inhibitor DDP_38003 (LSD1i) is able to penetrate the brain parenchyma in vivo in preclinical models, is well tolerated, and exerts antitumor activity in molecularly different GBMs. LSD1 genetic targeting further strengthens the role of LSD1 in GBM TIC maintenance. GBM TIC plasticity supports their adaptation and survival under a plethora of environmental stresses, including nutrient deficiency and proteostasis perturbation. By mimicking these stresses in vitro, we found that LSD1 inhibition hampers the induction of the activating transcription factor 4 (ATF4), the master regulator of the integrated stress response (ISR). The resulting aberrant ISR sensitizes GBM TICs to stress-induced cell death, hampering tumor aggressiveness. Functionally, LSD1i interferes with LSD1 scaffolding function and prevents its interaction with CREBBP, a critical ATF4 activator. By disrupting the interaction between CREBBP and LSD1-ATF4 axis, LSD1 inhibition prevents GBM TICs from overcoming stress and sustaining GBM progression. The effectiveness of the LSD1 inhibition in preclinical models shown here places a strong rationale toward its clinical translation for GBM treatment.
- Published
- 2021
- Full Text
- View/download PDF
26. Spatial epi-proteomics enabled by histone post-translational modification analysis from low-abundance clinical samples.
- Author
-
Noberini R, Savoia EO, Brandini S, Greco F, Marra F, Bertalot G, Pruneri G, McDonnell LA, and Bonaldi T
- Subjects
- Cell Line, Tumor drug effects, DNA Methylation, Histones genetics, Humans, Proteomics methods, Proteomics statistics & numerical data, Histones drug effects, Protein Processing, Post-Translational genetics
- Abstract
Background: Increasing evidence linking epigenetic mechanisms and different diseases, including cancer, has prompted in the last 15 years the investigation of histone post-translational modifications (PTMs) in clinical samples. Methods allowing the isolation of histones from patient samples followed by the accurate and comprehensive quantification of their PTMs by mass spectrometry (MS) have been developed. However, the applicability of these methods is limited by the requirement for substantial amounts of material., Results: To address this issue, in this study we streamlined the protein extraction procedure from low-amount clinical samples and tested and implemented different in-gel digestion strategies, obtaining a protocol that allows the MS-based analysis of the most common histone PTMs from laser microdissected tissue areas containing as low as 1000 cells, an amount approximately 500 times lower than what is required by available methods. We then applied this protocol to breast cancer patient laser microdissected tissues in two proof-of-concept experiments, identifying differences in histone marks in heterogeneous regions selected by either morphological evaluation or MALDI MS imaging., Conclusions: These results demonstrate that analyzing histone PTMs from very small tissue areas and detecting differences from adjacent tumor regions is technically feasible. Our method opens the way for spatial epi-proteomics, namely the investigation of epigenetic features in the context of tissue and tumor heterogeneity, which will be instrumental for the identification of novel epigenetic biomarkers and aberrant epigenetic mechanisms., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
27. Anticancer innovative therapy congress: Highlights from the 10th anniversary edition.
- Author
-
De Santis F, Fucà G, Schadendorf D, Mantovani A, Magnani L, Lisanti M, Pettitt S, Bellone M, Del Sal G, Minucci S, Eggermont A, Bruzzi P, Bicciato S, Conte P, Noberini R, Hiscott J, De Braud F, Del Vecchio M, and Di Nicola M
- Subjects
- Humans, Immunotherapy, Neoplasms drug therapy, Neoplastic Stem Cells, Anniversaries and Special Events, Therapies, Investigational
- Abstract
During the Tenth Edition of the Annual Congress on "Anticancer Innovative Therapy" [Milan, 23/24 January 2020], experts in the fields of immuno-oncology, epigenetics, tumor cell signaling, and cancer metabolism shared their latest knowledge on the roles of i] epigenetics, and in particular, chromatin modifiers, ii] cancer metabolism, iii] cancer stem cells [CSCs], iv] tumor cell signaling, and iv] the immune system. The novel therapeutic approaches presented included epigenetic drugs, cell cycle inhibitors combined with ICB, antibiotics and other off-label drugs, small-molecules active against CSCs, liposome-delivered miRNAs, tumor-specific CAR-T cells, and T-cell-based immunotherapy. Moreover, important evidence on possible mechanisms of resistance to these innovative therapies were also discussed, in particular with respect to resistance to ICB. Overall, this conference provided scientists and clinicians with a broad overview of future challenges and hopes to improve cancer treatment reasonably in the medium-short term., (Copyright © 2021. Published by Elsevier Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
28. Intestinal differentiation involves cleavage of histone H3 N-terminal tails by multiple proteases.
- Author
-
Ferrari KJ, Amato S, Noberini R, Toscani C, Fernández-Pérez D, Rossi A, Conforti P, Zanotti M, Bonaldi T, Tamburri S, and Pasini D
- Subjects
- Animals, Cathepsin L metabolism, Cell Differentiation, Homeostasis, Intestinal Mucosa cytology, Mice, Microvilli ultrastructure, Nucleosomes metabolism, Nucleosomes ultrastructure, Organoids, Protein Domains, Trypsin metabolism, Enterocytes metabolism, Histones metabolism, Intestine, Small cytology, Paneth Cells metabolism, Peptide Hydrolases metabolism, Protein Processing, Post-Translational, Stem Cells metabolism
- Abstract
The proteolytic cleavage of histone tails, also termed histone clipping, has been described as a mechanism for permanent removal of post-translational modifications (PTMs) from histone proteins. Such activity has been ascribed to ensure regulatory function in key cellular processes such as differentiation, senescence and transcriptional control, for which different histone-specific proteases have been described. However, all these studies were exclusively performed using cell lines cultured in vitro and no clear evidence that histone clipping is regulated in vivo has been reported. Here we show that histone H3 N-terminal tails undergo extensive cleavage in the differentiated cells of the villi in mouse intestinal epithelium. Combining biochemical methods, 3D organoid cultures and in vivo approaches, we demonstrate that intestinal H3 clipping is the result of multiple proteolytic activities. We identified Trypsins and Cathepsin L as specific H3 tail proteases active in small intestinal differentiated cells and showed that their proteolytic activity is differentially affected by the PTM pattern of histone H3 tails. Together, our findings provide in vivo evidence of H3 tail proteolysis in mammalian tissues, directly linking H3 clipping to cell differentiation., (© The Author(s) 2021. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2021
- Full Text
- View/download PDF
29. Enrichment of histones from patient samples for mass spectrometry-based analysis of post-translational modifications.
- Author
-
Noberini R, Restellini C, Savoia EO, and Bonaldi T
- Subjects
- Cells, Cultured, Female, Histones metabolism, Humans, Male, Methylation, Primary Cell Culture, Protein Processing, Post-Translational, Proteomics methods, Tandem Mass Spectrometry methods, Histocytological Preparation Techniques methods, Histones analysis, Specimen Handling methods
- Abstract
Aberrations in histone post-translational modifications (PTMs) have been implicated with the development of numerous pathologies, including cancer. Therefore, profiling histone PTMs in patient samples could provide information useful for the identification of epigenetic biomarkers, as well as for the discovery of potential novel targets. While antibody-based methods have been traditionally employed to analyze histone PTM in clinical samples, mass spectrometry (MS) can provide a more comprehensive, unbiased and quantitative view on histones and their PTMs. To combine the power of MS-based methods and the potential offered by histone PTM profiling of clinical samples, we have recently developed a series of methods for the extraction and enrichment of histones from different types of patient samples, including formalin-fixed paraffin-embedded tissues, fresh- and optimal cutting temperature-frozen tissues, and primary cells. Here, we provide a detailed description of these protocols, together with indications on the expected results and the most suitable workflow to be used downstream of each procedure., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
30. Label-Free Mass Spectrometry-Based Quantification of Linker Histone H1 Variants in Clinical Samples.
- Author
-
Noberini R, Morales Torres C, Savoia EO, Brandini S, Jodice MG, Bertalot G, Bonizzi G, Capra M, Diaferia G, Scaffidi P, and Bonaldi T
- Subjects
- Biomarkers, Tumor genetics, Epigenesis, Genetic genetics, Female, Gene Expression Regulation, Neoplastic genetics, Humans, Neoplasm Recurrence, Local diagnosis, Neoplasm Recurrence, Local pathology, Protein Processing, Post-Translational genetics, Tandem Mass Spectrometry, Triple Negative Breast Neoplasms diagnosis, Triple Negative Breast Neoplasms pathology, Histones genetics, Neoplasm Recurrence, Local genetics, Proteomics, Triple Negative Breast Neoplasms genetics
- Abstract
Epigenetic aberrations have been recognized as important contributors to cancer onset and development, and increasing evidence suggests that linker histone H1 variants may serve as biomarkers useful for patient stratification, as well as play an important role as drivers in cancer. Although traditionally histone H1 levels have been studied using antibody-based methods and RNA expression, these approaches suffer from limitations. Mass spectrometry (MS)-based proteomics represents the ideal tool to accurately quantify relative changes in protein abundance within complex samples. In this study, we used a label-free quantification approach to simultaneously analyze all somatic histone H1 variants in clinical samples and verified its applicability to laser micro-dissected tissue areas containing as low as 1000 cells. We then applied it to breast cancer patient samples, identifying differences in linker histone variants patters in primary triple-negative breast tumors with and without relapse after chemotherapy. This study highlights how label-free quantitation by MS is a valuable option to accurately quantitate histone H1 levels in different types of clinical samples, including very low-abundance patient tissues.
- Published
- 2020
- Full Text
- View/download PDF
31. Clinical Application of Mass Spectrometry-Based Proteomics in Lung Cancer Early Diagnosis.
- Author
-
Gasparri R, Sedda G, Noberini R, Bonaldi T, and Spaggiari L
- Subjects
- Biomarkers, Tumor metabolism, Humans, Early Detection of Cancer methods, Lung Neoplasms diagnosis, Lung Neoplasms metabolism, Mass Spectrometry, Proteomics
- Abstract
The current knowledge on proteomic biomarker analysis for the early diagnosis of lung cancer is summarized, underlining the diversity among the results and the current interest in translating research results into clinical practice. A MEDLINE/PubMed literature search to retrieve all the papers published in the last 10 years is performed. Proteomics studies on lung cancer have gathered evidence on the potential role of biomarkers in early diagnosis. Although promising, none of them have proved to be sufficiently reliable to achieve validation. Future research should evolve toward a multipanel analysis of proteins, considering the possibility that individual biomarkers might not be specific enough to diagnose lung cancer, but could be related to oncological conditions., (© 2020 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2020
- Full Text
- View/download PDF
32. hSWATH: Unlocking SWATH's Full Potential for an Untargeted Histone Perspective.
- Author
-
De Clerck L, Willems S, Noberini R, Restellini C, Van Puyvelde B, Daled S, Bonaldi T, Deforce D, and Dhaenens M
- Subjects
- Acetylation drug effects, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Line, Tumor, Female, Histone Deacetylase Inhibitors pharmacology, Humans, Peptide Library, Reproducibility of Results, Chromatography, Liquid methods, Histones metabolism, Peptides metabolism, Protein Processing, Post-Translational, Proteomics methods, Tandem Mass Spectrometry methods
- Abstract
Mass spectrometry (MS) has become the technique of choice for large-scale analysis of histone post-translational modifications (hPTMs) and their combinatorial patterns, especially in untargeted settings where novel discovery-driven hypotheses are being generated. However, MS-based histone analysis requires a distinct sample preparation, acquisition, and data analysis workflow when compared to traditional MS-based approaches. To this end, sequential window acquisition of all theoretical fragment ion spectra (SWATH) has great potential, as it allows for untargeted accurate identification and quantification of hPTMs. Here, we present a complete SWATH workflow specifically adapted for the untargeted study of histones (hSWATH). We assess its validity on a technical dataset of time-lapse deacetylation of a commercial histone extract using HDAC1, which contains a ground truth, i.e., acetylated substrate peptides reduce in intensity. We successfully apply this workflow in a biological setting and subsequently investigate the differential response to HDAC inhibition in different breast cancer cell lines.
- Published
- 2019
- Full Text
- View/download PDF
33. Epigenetic drug target deconvolution by mass spectrometry-based technologies.
- Author
-
Noberini R and Bonaldi T
- Subjects
- Animals, Humans, Protein Processing, Post-Translational drug effects, Proteome genetics, Proteome metabolism, Proteomics methods, Small Molecule Libraries chemistry, Drug Discovery methods, Epigenesis, Genetic drug effects, Mass Spectrometry methods, Small Molecule Libraries pharmacology
- Abstract
The identification of the full target spectrum of active molecules, known as target deconvolution, has become an indispensable step during the drug discovery process. It is now achievable thanks to mass spectrometry-based technologies. Here we discuss these approaches in the context of epigenetic drug discovery.
- Published
- 2019
- Full Text
- View/download PDF
34. Profiling of Epigenetic Features in Clinical Samples Reveals Novel Widespread Changes in Cancer.
- Author
-
Noberini R, Restellini C, Savoia EO, Raimondi F, Ghiani L, Jodice MG, Bertalot G, Bonizzi G, Capra M, Maffini FA, Tagliabue M, Ansarin M, Lupia M, Giordano M, Osti D, Pelicci G, Chiocca S, and Bonaldi T
- Abstract
Aberrations in histone post-translational modifications (PTMs), as well as in the histone modifying enzymes (HMEs) that catalyze their deposition and removal, have been reported in many tumors and many epigenetic inhibitors are currently under investigation for cancer treatment. Therefore, profiling epigenetic features in cancer could have important implications for the discovery of both biomarkers for patient stratification and novel epigenetic targets. In this study, we employed mass spectrometry-based approaches to comprehensively profile histone H3 PTMs in a panel of normal and tumoral tissues for different cancer types, identifying various changes, some of which appear to be a consequence of the increased proliferation rate of tumors, while others are cell-cycle independent. Histone PTM changes found in tumors partially correlate with alterations of the gene expression profiles of HMEs obtained from publicly available data and are generally lost in culture conditions. Through this analysis, we identified tumor- and subtype-specific histone PTM changes, but also widespread changes in the levels of histone H3 K9me3 and K14ac marks. In particular, H3K14ac showed a cell-cycle independent decrease in all the seven tumor/tumor subtype models tested and could represent a novel epigenetic hallmark of cancer. ., Competing Interests: The authors declare no conflict of interest
- Published
- 2019
- Full Text
- View/download PDF
35. Alternative digestion approaches improve histone modification mapping by mass spectrometry in clinical samples.
- Author
-
Restellini C, Cuomo A, Lupia M, Giordano M, Bonaldi T, and Noberini R
- Subjects
- Histone Code, Humans, Proteomics, Histones metabolism, Mass Spectrometry, Protein Processing, Post-Translational, Proteolysis
- Abstract
Purpose: Profiling histone posttranslational modifications (PTMs) in clinical samples holds great potential for the identification of epigenetic biomarkers and the discovery of novel epigenetic targets. MS-based approaches to analyze histone PTMs in clinical samples usually rely on SDS-PAGE separation following histone enrichment in order to eliminate detergents and further isolate histones. However, this limits the digestions options and hence the modification coverage., Experimental Design and Results: The aim of this study is the implementation of a procedure involving acetone protein precipitation followed by histone enrichment through a C18 StageTip column to obtain histone preparations suitable for various in-solution digestion protocols. Among them, the Arg-C digestion, which allows profiling histone H4 modifications, and the Prop-PIC method, which improves the detection of short and hydrophilic peptides, are tested. This approach is validated on different types of samples, including formalin-fixed paraffin-embedded pathology tissues, and employed to profile histone H4 modifications in cancer samples and normal tissues, identifying previously reported differences, as well as novel ones., Conclusions and Clinical Relevance: This protocol widens the number of applications available in the toolbox of clinical epigenomics, allowing the investigation of a larger spectrum of histone marks in patient samples., (© 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2019
- Full Text
- View/download PDF
36. Extensive and systematic rewiring of histone post-translational modifications in cancer model systems.
- Author
-
Noberini R, Osti D, Miccolo C, Richichi C, Lupia M, Corleone G, Hong SP, Colombo P, Pollo B, Fornasari L, Pruneri G, Magnani L, Cavallaro U, Chiocca S, Minucci S, Pelicci G, and Bonaldi T
- Subjects
- Animals, Breast Neoplasms genetics, Breast Neoplasms metabolism, Cell Line, Tumor, Epigenesis, Genetic, Female, Gene Expression Profiling, Glioblastoma genetics, Glioblastoma metabolism, Heterografts, Histones genetics, Humans, Mice, Mice, Nude, Models, Biological, Neoplasms genetics, Ovarian Neoplasms genetics, Ovarian Neoplasms metabolism, Protein Processing, Post-Translational, Proteomics, Tumor Cells, Cultured, Histone Code genetics, Histones metabolism, Neoplasms metabolism
- Abstract
Histone post-translational modifications (PTMs) generate a complex combinatorial code that regulates gene expression and nuclear functions, and whose deregulation has been documented in different types of cancers. Therefore, the availability of relevant culture models that can be manipulated and that retain the epigenetic features of the tissue of origin is absolutely crucial for studying the epigenetic mechanisms underlying cancer and testing epigenetic drugs. In this study, we took advantage of quantitative mass spectrometry to comprehensively profile histone PTMs in patient tumor tissues, primary cultures and cell lines from three representative tumor models, breast cancer, glioblastoma and ovarian cancer, revealing an extensive and systematic rewiring of histone marks in cell culture conditions, which includes a decrease of H3K27me2/me3, H3K79me1/me2 and H3K9ac/K14ac, and an increase of H3K36me1/me2. While some changes occur in short-term primary cultures, most of them are instead time-dependent and appear only in long-term cultures. Remarkably, such changes mostly revert in cell line- and primary cell-derived in vivo xenograft models. Taken together, these results support the use of xenografts as the most representative models of in vivo epigenetic processes, suggesting caution when using cultured cells, in particular cell lines and long-term primary cultures, for epigenetic investigations.
- Published
- 2018
- Full Text
- View/download PDF
37. PAT-H-MS coupled with laser microdissection to study histone post-translational modifications in selected cell populations from pathology samples.
- Author
-
Noberini R, Longuespée R, Richichi C, Pruneri G, Kriegsmann M, Pelicci G, and Bonaldi T
- Subjects
- Animals, Breast Neoplasms metabolism, Breast Neoplasms pathology, Carcinoma, Ductal, Breast metabolism, Carcinoma, Ductal, Breast pathology, Epigenesis, Genetic, Feasibility Studies, Female, Glioblastoma metabolism, Glioblastoma pathology, Humans, Leukemia metabolism, Leukemia pathology, Mice, Protein Processing, Post-Translational, Proteomics methods, Tissue Fixation, Histones metabolism, Laser Capture Microdissection methods, Mass Spectrometry methods, Neoplasms metabolism, Neoplasms pathology
- Abstract
Background: Aberrations in histone post-translational modifications (hPTMs) have been linked with various pathologies, including cancer, and could not only represent useful biomarkers but also suggest possible targetable epigenetic mechanisms. We have recently developed an approach, termed pathology tissue analysis of histones by mass spectrometry (PAT-H-MS), that allows performing a comprehensive and quantitative analysis of histone PTMs from formalin-fixed paraffin-embedded pathology samples. Despite its great potential, the application of this technique is limited by tissue heterogeneity., Methods: In this study, we further implemented the PAT-H-MS approach by coupling it with techniques aimed at reducing sample heterogeneity and selecting specific portions or cell populations within the samples, such as manual macrodissection and laser microdissection (LMD)., Results: When applied to the analysis of a small set of breast cancer samples, LMD-PAT-H-MS allowed detecting more marked changes between luminal A-like and triple negative patients as compared with the classical approach. These changes included not only the already known H3 K27me3 and K9me3 marks, but also H3 K36me1, which was found increased in triple negative samples and validated on a larger cohort of patients, and could represent a potential novel marker distinguishing breast cancer subtypes., Conclusions: These results show the feasibility of applying techniques to reduce sample heterogeneity, including laser microdissection, to the PAT-H-MS protocol, providing new tools in clinical epigenetics and opening new avenues for the comprehensive analysis of histone post-translational modifications in selected cell populations.
- Published
- 2017
- Full Text
- View/download PDF
38. A Super-SILAC Strategy for the Accurate and Multiplexed Profiling of Histone Posttranslational Modifications.
- Author
-
Noberini R and Bonaldi T
- Subjects
- Breast Neoplasms metabolism, Cell Line, Tumor, Chromatography, High Pressure Liquid, Female, Formaldehyde chemistry, Histones isolation & purification, Histones metabolism, Humans, Isotope Labeling, Paraffin Embedding, Protein Processing, Post-Translational, Proteomics, Tandem Mass Spectrometry, Tissue Fixation, Breast Neoplasms chemistry, Histones chemistry
- Abstract
Histone posttranslational modifications (hPTMs) generate a complex combinatorial code that plays a critical role in the regulation of gene activity and nuclear architecture during physiological and pathological processes. Mass spectrometry (MS) offers an unbiased, comprehensive, and quantitative view on hPTM patterns, and has emerged as a powerful tool in epigenetic research. Stable isotope labeling by amino acid in cell culture (SILAC) is a MS-based quantitative method that relies on the metabolic labeling of cell populations, which has been widely applied in global proteomic studies and can also be exploited for the accurate quantitation of hPTM changes among distinct functional states. However, the classical SILAC strategy has two main limits: it cannot be applied to more than three cell populations at the time and excludes samples that cannot be metabolically labeled, such as clinical samples. These limitations can be overcome by using a super-SILAC strategy, where a mix of heavy-labeled cell lines is used as a spike-in to analyze any types of samples with high accuracy and high multiplexing capabilities. In this chapter, we will provide a detailed description of a protocol to set up a histone-focused super-SILAC strategy and exploit it to accurately profile hPTMs across multiple samples. As a case study, we will describe a breast cancer-focused super-SILAC approach, which we used in a recent publication to profile hPTMs in frozen and formalin-fixed paraffin-embedded human samples, revealing previously unknown marks that differentiate breast cancer subtypes., (© 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
39. Protein kinase A can block EphA2 receptor-mediated cell repulsion by increasing EphA2 S897 phosphorylation.
- Author
-
Barquilla A, Lamberto I, Noberini R, Heynen-Genel S, Brill LM, and Pasquale EB
- Subjects
- Cell Line, Tumor, Cell Movement physiology, Cyclic AMP metabolism, Ephrin-A1 metabolism, Humans, Phosphorylation, Protein-Tyrosine Kinases, Proto-Oncogene Proteins c-akt metabolism, Receptors, Adrenergic metabolism, Serine metabolism, Signal Transduction, Small Molecule Libraries, Cyclic AMP-Dependent Protein Kinases metabolism, Receptor, EphA2 metabolism
- Abstract
The EphA2 receptor tyrosine kinase plays key roles in tissue homeostasis and disease processes such as cancer, pathological angiogenesis, and inflammation through two distinct signaling mechanisms. EphA2 "canonical" signaling involves ephrin-A ligand binding, tyrosine autophosphorylation, and kinase activity; EphA2 "noncanonical" signaling involves phosphorylation of serine 897 (S897) by AKT and RSK kinases. To identify small molecules counteracting EphA2 canonical signaling, we developed a high-content screening platform measuring inhibition of ephrin-A1-induced PC3 prostate cancer cell retraction. Surprisingly, most hits from a screened collection of pharmacologically active compounds are agents that elevate intracellular cAMP by activating G protein-coupled receptors such as the β2-adrenoceptor. We found that cAMP promotes phosphorylation of S897 by protein kinase A (PKA) as well as increases the phosphorylation of several nearby serine/threonine residues, which constitute a phosphorylation hotspot. Whereas EphA2 canonical and noncanonical signaling have been viewed as mutually exclusive, we show that S897 phosphorylation by PKA can coexist with EphA2 tyrosine phosphorylation and block cell retraction induced by EphA2 kinase activity. Our findings reveal a novel paradigm in EphA2 function involving the interplay of canonical and noncanonical signaling and highlight the ability of the β2-adrenoceptor/cAMP/PKA axis to rewire EphA2 signaling in a subset of cancer cells., (© 2016 Barquilla et al. This article is distributed by The American Society for Cell Biology under license from the author(s). Two months after publication it is available to the public under an Attribution–Noncommercial–Share Alike 3.0 Unported Creative Commons License (http://creativecommons.org/licenses/by-nc-sa/3.0).)
- Published
- 2016
- Full Text
- View/download PDF
40. Pathology Tissue-quantitative Mass Spectrometry Analysis to Profile Histone Post-translational Modification Patterns in Patient Samples.
- Author
-
Noberini R, Uggetti A, Pruneri G, Minucci S, and Bonaldi T
- Subjects
- Animals, Breast Neoplasms metabolism, Carcinoma, Ductal, Breast metabolism, Cell Line, Tumor, Female, Humans, Leukemia, Promyelocytic, Acute metabolism, Methylation, Mice, Proteomics methods, Tissue Fixation, Breast Neoplasms pathology, Carcinoma, Ductal, Breast pathology, Histones metabolism, Leukemia, Promyelocytic, Acute pathology, Protein Processing, Post-Translational, Tandem Mass Spectrometry methods
- Abstract
Histone post-translational modifications (hPTMs) generate a complex combinatorial code that has been implicated with various pathologies, including cancer. Dissecting such a code in physiological and diseased states may be exploited for epigenetic biomarker discovery, but hPTM analysis in clinical samples has been hindered by technical limitations. Here, we developed a method (PAThology tissue analysis of Histones by Mass Spectrometry - PAT-H-MS) that allows to perform a comprehensive, unbiased and quantitative MS-analysis of hPTM patterns on formalin-fixed paraffin-embedded (FFPE) samples. In pairwise comparisons, histone extracted from formalin-fixed paraffin-embedded tissues showed patterns similar to fresh frozen samples for 24 differentially modified peptides from histone H3. In addition, when coupled with a histone-focused version of the super-SILAC approach, this method allows the accurate quantification of modification changes among breast cancer patient samples. As an initial application of the PAThology tissue analysis of Histones by Mass Spectrometry method, we analyzed breast cancer samples, revealing significant changes in histone H3 methylation patterns among Luminal A-like and Triple Negative disease subtypes. These results pave the way for retrospective epigenetic studies that combine the power of MS-based hPTM analysis with the extensive clinical information associated with formalin-fixed paraffin-embedded archives., (© 2016 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
41. The contribution of mass spectrometry-based proteomics to understanding epigenetics.
- Author
-
Noberini R, Sigismondo G, and Bonaldi T
- Subjects
- Biomarkers chemistry, Chromatin metabolism, Histones analysis, Histones metabolism, Humans, Biomarkers analysis, Epigenesis, Genetic, Mass Spectrometry methods, Protein Processing, Post-Translational, Proteomics methods
- Abstract
Chromatin is a macromolecular complex composed of DNA and histones that regulate gene expression and nuclear architecture. The concerted action of DNA methylation, histone post-translational modifications and chromatin-associated proteins control the epigenetic regulation of the genome, ultimately determining cell fate and the transcriptional outputs of differentiated cells. Deregulation of this complex machinery leads to disease states, and exploiting epigenetic drugs is becoming increasingly attractive for therapeutic intervention. Mass spectrometry (MS)-based proteomics emerged as a powerful tool complementary to genomic approaches for epigenetic research, allowing the unbiased and comprehensive analysis of histone post-translational modifications and the characterization of chromatin constituents and chromatin-associated proteins. Furthermore, MS holds great promise for epigenetic biomarker discovery and represents a useful tool for deconvolution of epigenetic drug targets. Here, we will provide an overview of the applications of MS-based proteomics in various areas of chromatin biology.
- Published
- 2016
- Full Text
- View/download PDF
42. Mass-spectrometry analysis of histone post-translational modifications in pathology tissue using the PAT-H-MS approach.
- Author
-
Noberini R, Pruneri G, Minucci S, and Bonaldi T
- Abstract
Aberrant histone post-translational modifications (hPTMs) have been implicated with various pathologies, including cancer, and may represent useful epigenetic biomarkers. The data described here provide a mass spectrometry-based quantitative analysis of hPTMs from formalin-fixed paraffin-embedded (FFPE) tissues, from which histones were extracted through the recently developed PAT-H-MS method. First, we analyzed FFPE samples from mouse spleen and liver or human breast cancer up to six years old, together with their corresponding fresh frozen tissue. We then combined the PAT-H-MS approach with a histone-focused version of the super-SILAC strategy-using a mix of histones from four breast cancer cell lines as a spike-in standard- to accurately quantify hPTMs from breast cancer specimens belonging to different subtypes. The data, which are associated with a recent publication (Pathology tissue-quantitative mass spectrometry analysis to profile histone post-translational modification patterns in patient samples (Noberini, 2015) [1]), are deposited at the ProteomeXchange Consortium via the PRIDE partner repository with the dataset identifier PXD002669.
- Published
- 2016
- Full Text
- View/download PDF
43. Recent advances in mass spectrometry analysis of histone post-translational modifications: potential clinical impact of the PAT-H-MS approach.
- Author
-
Bonaldi T and Noberini R
- Subjects
- Female, Humans, Biomarkers, Tumor metabolism, Breast Neoplasms diagnosis, Histones metabolism, Mass Spectrometry methods, Protein Processing, Post-Translational
- Abstract
Histone post-translational modifications (hPTMs) contribute to the regulation of gene expression and increasing evidence links them to the development of various pathologies, highlighting their potential as biomarkers for prognostic, diagnostic and therapeutic applications. Mass spectrometry (MS) has emerged as a powerful analytical tool for hPTM analysis, which has also been applied to the analysis of epigenetic aberrations in diseases. However, the potential offered by the MS-based hPTM analysis of clinical samples for epigenetic biomarker discovery has been left largely unexploited. This article summarizes the contribution of MS-based approaches to clinical epigenetics, with a special focus on the PAThology tissue analysis of Histones by Mass Spectrometry (PAT-H-MS) approach--which represents the first application of MS-based hPTM analysis to formalin-fixed paraffin-embedded clinical samples--discussing its strengths and limitations, as well as possible implementations.
- Published
- 2016
- Full Text
- View/download PDF
44. Design, synthesis and bioevaluation of an EphA2 receptor-based targeted delivery system.
- Author
-
Barile E, Wang S, Das SK, Noberini R, Dahl R, Stebbins JL, Pasquale EB, Fisher PB, and Pellecchia M
- Subjects
- Amino Acid Sequence, Animals, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Cell Line, Tumor, Cell Survival drug effects, Drug Carriers chemistry, Female, Humans, Male, Mice, Mice, Inbred BALB C, Paclitaxel chemistry, Paclitaxel pharmacology, Paclitaxel therapeutic use, Peptides chemical synthesis, Peptides chemistry, Prostatic Neoplasms drug therapy, Receptor, EphA2 chemistry, Transplantation, Heterologous, Drug Carriers chemical synthesis, Drug Design, Receptor, EphA2 metabolism
- Abstract
Because of its overexpression in a range of solid tumors, the EphA2 receptor is a validated target for cancer therapeutics. We recently described a new targeted delivery system based on specific EphA2-targeting peptides conjugated with the chemotherapeutic agent paclitaxel. Here, we investigate the chemical determinants responsible for the stability and degradation of these agents in plasma. Introducing modifications in both the peptide and the linker between the peptide and paclitaxel resulted in drug conjugates that are both long-lived in rat plasma and that markedly decrease tumor size in a prostate cancer xenograft model compared with paclitaxel alone treatment. These studies identify critical rate-limiting degradation sites on the peptide-drug conjugates, enabling the design of agents with increased stability and efficacy. These results provide support for our central hypothesis that peptide-drug conjugates targeting EphA2 represent an innovative and potentially effective strategy to selectively deliver cytotoxic drugs to cancer cells., (© 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2014
- Full Text
- View/download PDF
45. Quantitative chemical proteomics identifies novel targets of the anti-cancer multi-kinase inhibitor E-3810.
- Author
-
Colzani M, Noberini R, Romanenghi M, Colella G, Pasi M, Fancelli D, Varasi M, Minucci S, and Bonaldi T
- Subjects
- Humans, Isotope Labeling, Mass Spectrometry, Molecular Targeted Therapy, Neoplasm Proteins antagonists & inhibitors, Receptor-Interacting Protein Serine-Threonine Kinase 2 antagonists & inhibitors, Vascular Endothelial Growth Factor A metabolism, Naphthalenes administration & dosage, Neoplasm Proteins biosynthesis, Protein Kinase Inhibitors administration & dosage, Proteomics, Quinolines administration & dosage, Vascular Endothelial Growth Factor A antagonists & inhibitors
- Abstract
Novel drugs are designed against specific molecular targets, but almost unavoidably they bind non-targets, which can cause additional biological effects that may result in increased activity or, more frequently, undesired toxicity. Chemical proteomics is an ideal approach for the systematic identification of drug targets and off-targets, allowing unbiased screening of candidate interactors in their natural context (tissue or cell extracts). E-3810 is a novel multi-kinase inhibitor currently in clinical trials for its anti-angiogenic and anti-tumor activity. In biochemical assays, E-3810 targets primarily vascular endothelial growth factor and fibroblast growth factor receptors. Interestingly, E-3810 appears to inhibit the growth of tumor cells with low to undetectable levels of these proteins in vitro, suggesting that additional relevant targets exist. We applied chemical proteomics to screen for E-3810 targets by immobilizing the drug on a resin and exploiting stable isotope labeling by amino acids in cell culture to design experiments that allowed the detection of novel interactors and the quantification of their dissociation constant (Kd imm) for the immobilized drug. In addition to the known target FGFR2 and PDGFRα, which has been described as a secondary E-3810 target based on in vitro assays, we identified six novel candidate kinase targets (DDR2, YES, LYN, CARDIAK, EPHA2, and CSBP). These kinases were validated in a biochemical assay and-in the case of the cell-surface receptor DDR2, for which activating mutations have been recently discovered in lung cancer-cellular assays. Taken together, the success of our strategy-which integrates large-scale target identification and quality-controlled target affinity measurements using quantitative mass spectrometry-in identifying novel E-3810 targets further supports the use of chemical proteomics to dissect the mechanism of action of novel drugs., (© 2014 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
46. Amino acid conjugates of lithocholic acid as antagonists of the EphA2 receptor.
- Author
-
Incerti M, Tognolini M, Russo S, Pala D, Giorgio C, Hassan-Mohamed I, Noberini R, Pasquale EB, Vicini P, Piersanti S, Rivara S, Barocelli E, Mor M, and Lodola A
- Subjects
- Adenocarcinoma pathology, Enzyme-Linked Immunosorbent Assay, Humans, Lithocholic Acid chemistry, Male, Models, Molecular, Phosphorylation, Prostatic Neoplasms pathology, Receptor, EphA2 metabolism, Amino Acids chemistry, Lithocholic Acid pharmacology, Receptor, EphA2 antagonists & inhibitors
- Abstract
The Eph receptor-ephrin system is an emerging target for the development of novel antiangiogenetic agents. We recently identified lithocholic acid (LCA) as a small molecule able to block EphA2-dependent signals in cancer cells, suggesting that its (5β)-cholan-24-oic acid scaffold can be used as a template to design a new generation of improved EphA2 antagonists. Here, we report the design and synthesis of an extended set of LCA derivatives obtained by conjugation of its carboxyl group with different α-amino acids. Structure-activity relationships indicate that the presence of a lipophilic amino acid side chain is fundamental to achieve good potencies. The l-Trp derivative (20, PCM126) was the most potent antagonist of the series disrupting EphA2-ephrinA1 interaction and blocking EphA2 phosphorylation in prostate cancer cells at low μM concentrations, thus being significantly more potent than LCA. Compound 20 is among the most potent small-molecule antagonists of the EphA2 receptor.
- Published
- 2013
- Full Text
- View/download PDF
47. Design, synthesis and characterization of novel small molecular inhibitors of ephrin-B2 binding to EphB4.
- Author
-
Duggineni S, Mitra S, Noberini R, Han X, Lin N, Xu Y, Tian W, An J, Pasquale EB, and Huang Z
- Subjects
- Amino Acid Sequence, Computer Simulation, Ephrin-B2 metabolism, Models, Molecular, Protein Binding, Protein Conformation, Receptor, EphB4 genetics, Signal Transduction, Antineoplastic Agents pharmacology, Drug Design, Ephrin-B2 antagonists & inhibitors, Receptor, EphB4 metabolism
- Abstract
EphB4 is a member of the large Eph receptor tyrosine kinase family. By interacting with its preferred ligand ephrin-B2, which is also a transmembrane protein, EphB4 plays a role in a variety of physiological and pathological processes ranging from bone remodeling to cancer malignancy. EphB4-ephrin-B2 binding occurs at sites of contact between cells. Ephrin-B2 causes EphB4 clustering and increased kinase activity to generate downstream signals that affect cell behavior. Previous work identified a high-affinity antagonistic peptide that targets EphB4, named TNYL-RAW. This peptide is 15 amino acid long, has a molecular weight of ~1700 Da and binds to the ephrin-binding pocket of EphB4. Here we report the structure-based design and chemical synthesis of two novel small molecules of ~600-700 Da, which were designed starting from the small and functionally critical C-terminal portion of the TNYL-RAW peptide. These compounds inhibit ephrin-B2 binding to EphB4 at low micromolar concentrations. Additionally, although the ephrin-B2 ligand can interacts with multiple other Eph receptors besides EphB4, the two compounds retain the high selectivity of the TNYL-RAW peptide in targeting EphB4. TNYL-RAW peptide displacement experiments using the more potent of the two compounds, compound 5, suggest a competitive mode of inhibition. These EphB4 antagonistic compounds can serve as promising templates for the further development of small molecule drugs targeting EphB4., (Published by Elsevier Inc.)
- Published
- 2013
- Full Text
- View/download PDF
48. HTS by NMR of combinatorial libraries: a fragment-based approach to ligand discovery.
- Author
-
Wu B, Zhang Z, Noberini R, Barile E, Giulianotti M, Pinilla C, Houghten RA, Pasquale EB, and Pellecchia M
- Subjects
- High-Throughput Screening Assays methods, Humans, Ligands, Models, Molecular, Protein Structure, Tertiary, Receptor, EphA4 chemistry, Structure-Activity Relationship, X-Linked Inhibitor of Apoptosis Protein chemistry, X-Linked Inhibitor of Apoptosis Protein metabolism, Combinatorial Chemistry Techniques methods, Drug Design, Magnetic Resonance Spectroscopy methods, Receptor, EphA4 metabolism, Small Molecule Libraries chemistry, Small Molecule Libraries pharmacology
- Abstract
Fragment-based ligand design (FBLD) approaches have become more widely used in drug discovery projects from both academia and industry, and are even often preferred to traditional high-throughput screening (HTS) of large collection of compounds (>10(5)). A key advantage of FBLD approaches is that these often rely on robust biophysical methods such as NMR spectroscopy for detection of ligand binding, hence are less prone to artifacts that too often plague the results from HTS campaigns. In this article, we introduce a screening strategy that takes advantage of both the robustness of protein NMR spectroscopy as the detection method, and the basic principles of combinatorial chemistry to enable the screening of large libraries of fragments (>10(5) compounds) preassembled on a common backbone. We used the method to identify compounds that target protein-protein interactions., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
49. Targeted delivery of paclitaxel to EphA2-expressing cancer cells.
- Author
-
Wang S, Noberini R, Stebbins JL, Das S, Zhang Z, Wu B, Mitra S, Billet S, Fernandez A, Bhowmick NA, Kitada S, Pasquale EB, Fisher PB, and Pellecchia M
- Subjects
- Animals, Antineoplastic Agents, Phytogenic chemistry, Disease Models, Animal, Female, Gene Expression, Humans, Male, Mice, Neoplasms drug therapy, Neoplasms pathology, Neovascularization, Pathologic drug therapy, Neovascularization, Pathologic genetics, Paclitaxel chemistry, Receptor, EphA2 metabolism, Transplantation, Homologous, Tumor Burden drug effects, Xenograft Model Antitumor Assays, Antineoplastic Agents, Phytogenic administration & dosage, Drug Delivery Systems, Neoplasms genetics, Paclitaxel administration & dosage, Peptides chemistry, Receptor, EphA2 genetics
- Abstract
Purpose: YSA is an EphA2-targeting peptide that effectively delivers anticancer agents to prostate cancer tumors. Here, we report on how we increased the drug-like properties of this delivery system., Experimental Design: By introducing non-natural amino acids, we have designed two new EphA2 targeting peptides: YNH, where norleucine and homoserine replace the two methionine residues of YSA, and dYNH, where a D-tyrosine replaces the L-tyrosine at the first position of the YNH peptide. We describe the details of the synthesis of YNH and dYNH paclitaxel conjugates (YNH-PTX and dYNH-PTX) and their characterization in cells and in vivo., Results: dYNH-PTX showed improved stability in mouse serum and significantly reduced tumor size in a prostate cancer xenograft model and also reduced tumor vasculature in a syngeneic orthotopic allograft mouse model of renal cancer compared with vehicle or paclitaxel treatments., Conclusion: This study reveals that targeting EphA2 with dYNH drug conjugates could represent an effective way to deliver anticancer agents to a variety of tumor types.
- Published
- 2013
- Full Text
- View/download PDF
50. Inhibition of Eph receptor-ephrin ligand interaction by tea polyphenols.
- Author
-
Noberini R, Koolpe M, Lamberto I, and Pasquale EB
- Subjects
- Animals, COS Cells, Catechin analogs & derivatives, Catechin chemistry, Catechin pharmacology, Cell Line, Chlorocebus aethiops, Mice, Protein Binding drug effects, Receptor, EphA4 antagonists & inhibitors, Receptor, EphA4 metabolism, Receptors, Eph Family antagonists & inhibitors, Signal Transduction drug effects, Small Molecule Libraries chemistry, Small Molecule Libraries pharmacology, Ephrins metabolism, Polyphenols chemistry, Polyphenols pharmacology, Protein Interaction Maps drug effects, Receptors, Eph Family metabolism, Tea chemistry
- Abstract
Tea contains a variety of bioactive chemicals, such as catechins and other polyphenols. These compounds are thought to be responsible for the health benefits of tea consumption by affecting the function of many cellular targets, not all of which have been identified. In a high-throughput screen for small molecule antagonists of the EphA4 receptor tyrosine kinase, we identified five tea polyphenols that substantially inhibit EphA4 binding to a synthetic peptide ligand. Further characterization of theaflavin monogallates from black tea and epigallocatechin-3,5-digallate from green tea revealed that these compounds at low micromolar concentrations also inhibit binding of the natural ephrin ligands to EphA4 and several other Eph receptors in in vitro assays. The compounds behave as competitive EphA4 antagonists, and their inhibitory activity is affected by amino acid mutations within the ephrin binding pocket of EphA4. In contrast, the major green tea catechin, epigallocatechin-3-gallate (EGCG), does not appear to be an effective Eph receptor antagonist. In cell culture assays, theaflavin monogallates and epigallocatechin-3,5-digallate inhibit ephrin-induced tyrosine phosphorylation (activation) of Eph receptors and endothelial capillary-like tube formation. However, the wider spectrum of Eph receptors affected by the tea derivatives in cells suggests additional mechanisms of inhibition besides interfering with ephrin binding. These results show that tea polyphenols derived from both black and green tea can suppress the biological activities of Eph receptors. Thus, the Eph receptor tyrosine kinase family represents an important class of targets for tea-derived phytochemicals., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.