28 results on '"Alba Maiques-Diaz"'
Search Results
2. S142: FOXO1-RICTOR AXIS INDUCES ADAPTIVE INCREASE IN AKT ACTIVITY DURING BCR INHIBITOR THERAPY IN CLL: IMPLICATIONS FOR COMBINATION THERAPY
- Author
-
Laura Ondrisova, Vaclav Seda, Eva Hoferkova, Giorgia Chiodin, Krystof Hlavac, Lenka Kostalova, Daniel Filip, Pedro Faria Zeni, Anna Panovska, Karla Plevova, Sarka Pospisilova, Martin Simkovic, Filip Vrbacky, Daniel Lysak, Stacey M. Fernandes, Matthew S. Davids, Alba Maiques-Diaz, Stella Charalampopoulou, Jose I Martin-Subero, Jennifer R Brown, Michael Doubek, Francesco Forconi, Jiri Mayer, and Marek Mraz
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
3. Robust CRISPR-Cas9 Genetic Editing of Primary Chronic Lymphocytic Leukemia and Mantle Cell Lymphoma Cells
- Author
-
Judith Mateos-Jaimez, Maurizio Mangolini, Anna Vidal, Marta Kulis, Dolors Colomer, Elias Campo, Ingo Ringshausen, Jose I. Martin-Subero, and Alba Maiques-Diaz
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
4. 'Long COVID-19' of Researchers: What to Do Next?
- Author
-
Eleni Gavriilaki and Alba Maiques-Diaz
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2022
- Full Text
- View/download PDF
5. Effect of the COVID-19 Pandemic on Laboratory and Clinical Research: A Testimony and a Call to Action From Researchers
- Author
-
Anna Kabanova, Eleni Gavriilaki, Benedikt W. Pelzer, Lorenzo Brunetti, and Alba Maiques-Diaz
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2020
- Full Text
- View/download PDF
6. Enhancer Activation by Pharmacologic Displacement of LSD1 from GFI1 Induces Differentiation in Acute Myeloid Leukemia
- Author
-
Alba Maiques-Diaz, Gary J. Spencer, James T. Lynch, Filippo Ciceri, Emma L. Williams, Fabio M.R. Amaral, Daniel H. Wiseman, William J. Harris, Yaoyong Li, Sudhakar Sahoo, James R. Hitchin, Daniel P. Mould, Emma E. Fairweather, Bohdan Waszkowycz, Allan M. Jordan, Duncan L. Smith, and Tim C.P. Somervaille
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Pharmacologic inhibition of LSD1 promotes blast cell differentiation in acute myeloid leukemia (AML) with MLL translocations. The assumption has been that differentiation is induced through blockade of LSD1’s histone demethylase activity. However, we observed that rapid, extensive, drug-induced changes in transcription occurred without genome-wide accumulation of the histone modifications targeted for demethylation by LSD1 at sites of LSD1 binding and that a demethylase-defective mutant rescued LSD1 knockdown AML cells as efficiently as wild-type protein. Rather, LSD1 inhibitors disrupt the interaction of LSD1 and RCOR1 with the SNAG-domain transcription repressor GFI1, which is bound to a discrete set of enhancers located close to transcription factor genes that regulate myeloid differentiation. Physical separation of LSD1/RCOR1 from GFI1 is required for drug-induced differentiation. The consequent inactivation of GFI1 leads to increased enhancer histone acetylation within hours, which directly correlates with the upregulation of nearby subordinate genes. : Maiques-Diaz et al. report that, while LSD1 inhibitors target both scaffolding and enzymatic functions of the protein, drug-induced myeloid leukemia cell differentiation is primarily due to the disruption and release from enhancers of GFI1/CoREST complexes, leading to the activation of subordinate myeloid transcription factor genes. Keywords: LSD1, GFI1, acute myeloid leukemia, MLL, acetylation, methylation
- Published
- 2018
- Full Text
- View/download PDF
7. LSD1 inhibitors disrupt the GFI1 transcription repressor complex
- Author
-
Alba Maiques-Diaz, James T. Lynch, Gary J. Spencer, and Tim C.P. Somervaille
- Subjects
gfi1 ,lsd1 ,aml ,epigenetics ,methylation ,acetylation ,leukemia ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Pharmacologic inhibition of KDM1A (Lysine Demethylase 1A) induces differentiation in certain subtypes of acute myeloid leukemia. Our recent studies reveal this is dependent upon drug-induced disruption of the GFI1 (Growth Factor Independent 1) transcription repressor complex, leading to activation of enhancers distributed close to genes controlling monocytic lineage differentiation.
- Published
- 2018
- Full Text
- View/download PDF
8. Abrogation of RUNX1 gene expression in de novo myelodysplastic syndrome with t(4;21)(q21;q22)
- Author
-
Ana Rio-Machín, Juliane Menezes, Alba Maiques-Diaz, Xabier Agirre, Bibiana I. Ferreira, Francesco Acquadro, Sandra Rodriguez-Perales, Karmele A. Juaristi, Sara Álvarez, and Juan C. Cigudosa
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
The disruption of RUNX1 function is one of the main mechanisms of disease observed in hematopoietic malignancies and the description of novel genetic events that lead to a RUNX1 loss of function has been accelerated with the development of genomic technologies. Here we describe the molecular characterization of a new t(4;21)(q21;q22) in a de novo myelodysplastic syndrome that resulted in the deletion of the RUNX1 gene. We demonstrated by quantitative real-time RT-PCR an almost complete depletion of the expression of the RUNX1 gene in our t(4;21) case compared with CD34+ cells that was independent of mutation or DNA methylation. More importantly, we explored and confirmed the possibility that this abrogation also prevented transactivation of RUNX1 target genes, perhaps confirming the genetic origin of the thrombocytopenia and the myelodysplastic features observed in our patient, and certainly mimicking what has been observed in the presence of the RUNX1/ETO fusion protein.
- Published
- 2012
- Full Text
- View/download PDF
9. HMG20B stabilizes association of LSD1 with GFI1 on chromatin to confer transcription repression and leukemia cell differentiation block
- Author
-
Alba Maiques-Diaz, Luciano Nicosia, Naseer J. Basma, Isabel Romero-Camarero, Francesco Camera, Gary J. Spencer, Fabio M. R. Amaral, Fabrizio Simeoni, Bettina Wingelhofer, Andrew J. K. Williamson, Andrew Pierce, Anthony D. Whetton, and Tim C. P. Somervaille
- Subjects
Cancer Research ,Genetics ,Molecular Biology - Abstract
Pharmacologic inhibition of LSD1 induces molecular and morphologic differentiation of blast cells in acute myeloid leukemia (AML) patients harboring MLL gene translocations. In addition to its demethylase activity, LSD1 has a critical scaffolding function at genomic sites occupied by the SNAG domain transcription repressor GFI1. Importantly, inhibitors block both enzymatic and scaffolding activities, in the latter case by disrupting the protein:protein interaction of GFI1 with LSD1. To explore the wider consequences of LSD1 inhibition on the LSD1 protein complex we applied mass spectrometry technologies. We discovered that the interaction of the HMG-box protein HMG20B with LSD1 was also disrupted by LSD1 inhibition. Downstream investigations revealed that HMG20B is co-located on chromatin with GFI1 and LSD1 genome-wide; the strongest HMG20B binding co-locates with the strongest GFI1 and LSD1 binding. Functional assays demonstrated that HMG20B depletion induces leukemia cell differentiation and further revealed that HMG20B is required for the transcription repressor activity of GFI1 through stabilizing LSD1 on chromatin at GFI1 binding sites. Interaction of HMG20B with LSD1 is through its coiled-coil domain. Thus, HMG20B is a critical component of the GFI1:LSD1 transcription repressor complex which contributes to leukemia cell differentiation block.
- Published
- 2022
10. Therapeutic Targeting of EP300/CBP By Bromodomain Inhibition in Acute Myeloid Leukemia
- Author
-
Luciano Nicosia, Gary J Spencer, Nigel Brooks, Fabio Amaral, Naseer Basma, John Chadwick, Bradley Revell, Bettina Wingelhofer, Alba Maiques-Diaz, Filippo Ciceri, Daniel H Wiseman, Neil A Pegg, William West, Tomasz Knurowski, Kris Frese, Karen Clegg, Victoria Louise Campbell, Mhairi Copland, Emma Searle, and Tim C.P Somervaille
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
11. Development of a Novel Chromatin Activation Score As a Powerful Independent Prognostic Factor in Chronic Lymphocytic Leukemia
- Author
-
Vicente Chapaprieta, Alba Maiques-Diaz, Guillem Clot, Ferran Nadeu, Judith Mateos-Jaimez, Anna Vidal, Stella Charalampopoulou, Martí Duran-Ferrer, Romina Royo, Junyan Lu, Thorsten Zenz, Julio Delgado, Armando Lopez-Guillermo, Elías Campo, and José I. Martín-Subero
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
12. CRISPR-Cas9 Genome Editing of Primary Chronic Lymphocytic Leukemia Cells
- Author
-
Judith Mateos-Jaimez, Maurizio Mangolini, Anna Vidal, Dolors Colomer, Elías Campo, Ingo Ringshausen, José I. Martín-Subero, and Alba Maiques-Diaz
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
13. 'Long COVID-19' of Researchers: What to Do Next?
- Author
-
Eleni Gavriilaki and Alba Maiques-Diaz
- Subjects
Perspective ,Diseases of the blood and blood-forming organs ,Hematology ,RC633-647.5 - Published
- 2021
14. Development of (4-Cyanophenyl)glycine Derivatives as Reversible Inhibitors of Lysine Specific Demethylase 1
- Author
-
Gary J. Spencer, Ulf Bremberg, Matthis Geitmann, Allan M. Jordan, Sharon Cartic, Alison E. McGonagle, Daniel P. Mould, Fabrice Turlais, Donald J. Ogilvie, Tim C. P. Somervaille, Cristina Alli, and Alba Maiques-Diaz
- Subjects
Models, Molecular ,0301 basic medicine ,In silico ,hERG ,Glycine ,Antineoplastic Agents ,Pharmacology ,Compound 32 ,Structure-Activity Relationship ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Drug Discovery ,medicine ,Humans ,Structure–activity relationship ,Computer Simulation ,Spiro Compounds ,Enzyme Inhibitors ,Histone Demethylases ,Leukemia ,Manchester Cancer Research Centre ,biology ,Drug discovery ,Chemistry ,ResearchInstitutes_Networks_Beacons/mcrc ,Tranylcypromine ,Myeloid leukemia ,medicine.disease ,Ether-A-Go-Go Potassium Channels ,High-Throughput Screening Assays ,Molecular Docking Simulation ,030104 developmental biology ,Biochemistry ,Drug Design ,030220 oncology & carcinogenesis ,biology.protein ,Molecular Medicine ,Biomarkers ,medicine.drug - Abstract
Inhibition of lysine specific demethylase 1 (LSD1) has been shown to induce the differentiation of leukemia stem cells in acute myeloid leukaemia (AML). Irreversible inhibitors developed from the non-specific inhibitor tranylcypromine have entered clinical trials; however, the development of effective reversible inhibitors has proved more challenging. Herein, we describe our efforts to identify reversible inhibitors of LSD1 from a high throughput screen, and subsequent in silico modelling approaches. From a single hit (12) validated by biochemical and biophysical assays, we describe our efforts to develop acyclic scaffold-hops from GSK-690 (1). A further scaffold modification to a (4-cyanophenyl)glycinamide (e.g. 29a) led to the development of compound 32, with a Kd value of 32 nM and an EC50 value of 0.67 μM in a surrogate cellular biomarker assay. Moreover, this derivative does not display the same level of hERG liability as observed with 1 and represents a promising lead for further development.
- Published
- 2017
15. Pre-clinical activity of combined LSD1 and mTORC1 inhibition in MLL-translocated acute myeloid leukaemia
- Author
-
Bettina Wingelhofer, Gauri Deb, Alba Maiques-Diaz, John A. Chadwick, Tim C. P. Somervaille, Emma Williams, Fabio M. R. Amaral, Tamara Maes, Gary J. Spencer, and Hui-Sun Leong
- Subjects
0301 basic medicine ,Cancer Research ,Myeloid ,Apoptosis ,Mice, SCID ,Translocation, Genetic ,Mice ,0302 clinical medicine ,Mice, Inbred NOD ,hemic and lymphatic diseases ,Tumor Cells, Cultured ,Histone Demethylases ,Gene knockdown ,Manchester Cancer Research Centre ,Gene Expression Regulation, Leukemic ,Hematology ,Antidepressive Agents ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Drug Therapy, Combination ,Female ,Myeloid-Lymphoid Leukemia Protein ,Antineoplastic Agents ,Biology ,Mechanistic Target of Rapamycin Complex 1 ,Article ,Acute myeloid leukaemia ,03 medical and health sciences ,Downregulation and upregulation ,medicine ,Animals ,Humans ,Chemotherapy ,Everolimus ,Clonogenic assay ,Cell Proliferation ,ResearchInstitutes_Networks_Beacons/mcrc ,KDM1A ,Histone-Lysine N-Methyltransferase ,medicine.disease ,Minimal residual disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Cancer research ,biology.protein ,Demethylase ,Tranylcypromine - Abstract
The histone demethylase lysine-specific demethylase 1 (LSD1 or KDM1A) has emerged as a candidate therapeutic target in acute myeloid leukaemia (AML); tranylcypromine-derivative inhibitors induce loss of clonogenic activity and promote differentiation, in particular in the MLL-translocated molecular subtype of AML. In AML, the use of drugs in combination often delivers superior clinical activity. To identify genes and cellular pathways that collaborate with LSD1 to maintain the leukaemic phenotype, and which could be targeted by combination therapies, we performed a genome-wide CRISPR-Cas9 dropout screen. We identified multiple components of the amino acid sensing arm of mTORC1 signalling—RRAGA, MLST8, WDR24 and LAMTOR2—as cellular sensitizers to LSD1 inhibition. Knockdown of mTORC1 components, or mTORC1 pharmacologic inhibition, in combination with LSD1 inhibition enhanced differentiation in both cell line and primary cell settings, in vitro and in vivo, and substantially reduced the frequency of clonogenic primary human AML cells in a modelled minimal residual disease setting. Synergistic upregulation of a set of transcription factor genes associated with terminal monocytic lineage differentiation was observed. Thus, dual mTORC1 and LSD1 inhibition represents a candidate combination approach for enhanced differentiation in MLL-translocated AML which could be evaluated in early phase clinical trials.
- Published
- 2019
16. LSD1: biologic roles and therapeutic targeting
- Author
-
Alba Maiques-Diaz and Tim C. P. Somervaille
- Subjects
0301 basic medicine ,Cancer Research ,ORY1001 ,Regulator ,LSD1 ,Antineoplastic Agents ,Review ,acute myeloid leukemia ,Pharmacology ,Biology ,Therapeutic targeting ,Bioinformatics ,Epigenesis, Genetic ,03 medical and health sciences ,histone demethylase ,Journal Article ,small-cell lung cancer ,Genetics ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Epigenetics ,Enzyme Inhibitors ,Histone demethylase activity ,GSK2879552 ,Histone Demethylases ,Manchester Cancer Research Centre ,ResearchInstitutes_Networks_Beacons/mcrc ,Tranylcypromine ,KDM1A ,Gene Expression Regulation, Neoplastic ,Clinical trial ,Leukemia, Myeloid, Acute ,030104 developmental biology ,medicine.drug - Abstract
LSD1 (KDM1A; BHC110; AOF2) was the first protein reported to exhibit histone demethylase activity and has since been shown to have multiple essential roles in mammalian biology. Given its enzymatic activity and its high-level expression in many human malignancies, a significant recent focus has been the development of pharmacologic inhibitors. Here we summarize structural and biochemical knowledge of this important epigenetic regulator, with a particular emphasis on the functional and preclinical studies in oncology that have provided justification for the evaluation of tranylcypromine derivative LSD1 inhibitors in early phase clinical trials.
- Published
- 2016
17. Effect of the COVID-19 Pandemic on Laboratory and Clinical Research: A Testimony and a Call to Action From Researchers
- Author
-
Alba Maiques-Diaz, Lorenzo Brunetti, Anna Kabanova, Benedikt W. Pelzer, and Eleni Gavriilaki
- Subjects
2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,lcsh:RC633-647.5 ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,MEDLINE ,lcsh:Diseases of the blood and blood-forming organs ,Hematology ,medicine.disease ,Call to action ,Clinical research ,Perspective ,Pandemic ,medicine ,Medical emergency ,Psychology - Published
- 2020
18. MAPK8-mediated stabilization of SP1 is essential for RUNX1-RUNX1T1 - driven leukaemia
- Author
-
Sara Alvarez, Mahesh Shrestha, Ana Rio-Machin, Miriam Hernando, Juan C. Cigudosa, Alba Maiques-Diaz, Amanda Sánchez-López, and James C. Mulloy
- Subjects
0301 basic medicine ,Oncogene Proteins, Fusion ,Sp1 Transcription Factor ,Chemistry ,MAPK8 ,Hematology ,Leukemia, Myeloid, Acute ,03 medical and health sciences ,RUNX1 Translocation Partner 1 Protein ,030104 developmental biology ,Cell Line, Tumor ,Core Binding Factor Alpha 2 Subunit ,Runx1 runx1t1 ,Cancer research ,Humans ,Mitogen-Activated Protein Kinase 8 ,Myeloid leukaemia - Published
- 2015
19. Abstracts from the 4th World Congress of the International Dermoscopy Society, April 16-18, 2015, Vienna, Austria
- Author
-
Michael A. Marchetti, Alexandros Stratigos, Claudia Jaeger, Nanja van Geel, Erika Varga, Rachel M Bowden, Nebojsa Pesic, Lauren A. Penn, Francesca Farnetani, Irena Walecka, Otto S. Wolfbeis, Anna Pogorzelska-Antkowiak, Małgorzata Zadurska, Miriam A. Jesús Silva, Mari Grönroos, Fabrizio Ayala, Claudia Sprincenatu, Ausilia Maria Manganoni, Jhonatan Rafael S. Pinheiro, Vincent Descamps, Era C. Murzaku, Josephine Rau, Christian Landi, Josep Malvehy, Othon Papadopoulos, Renato Talamini, Savitha L. Beergouder, Adrian Ballano Ruiz, Karina Scandura, Flavia Persechino, Yunxian Tian, Mark Berneburg, Iara Drakensjö, Luis Javier Del pozo, Elizabeth Lazaridou, Marwah A. Saleh, Wei Zhang, Dalal Mosaad, Aida Carolina Medina, Alka Lalji, Robabeh Abedini, FZ Debagh, Ligia Brzezinska-Wcislo, Nurşah Doğan, Naglaa Ahmed, Tamerlan Shaipov, Ritta Khoury, Lidija Kandolf-Sekulovic, Aldo Bono, Luis Angel Vera, Naotomo Kambe, Jaka Rados, Sergio Talarico, Milvia Maria S. E. S. Enokihara, Iris Zalaudek, Malgorzata Maj, Francesca Specchio, Paloma Arribas, Nazan Emiroglu, Andreea Ioana Popescu, Irina Sergeeva, Virginia Chitu, Michael Kirschbaum, Sergio Yamada, Niken Wulandari, Rotaru Maria, Lore Pil, Lieve Brochez, Anthony Azzi, Vasiliy Y. Sergeev, Raimonds Karls, Zeynep Topkarci, Tanja Planinsek Rucigaj, Osvania Maris, Graham J. Mann, Timótio Dorn, Lubomir Drlik, Pilar Iranzo, Sara Minghetti, Michael Noe, Ahmet R Akar, Jesus Cuevas Santos, Laura Raducu, Salim Ysmail-Dahlouk, Laura Mazzoni, Sidharth Sonthalia, Neşe Çallı Demirkan, Yaei Togawa, Branislava Gajic, Ayelet Rishpon, Chih-Hsun Yang, Barbara Boone, José Luis López-Estebaranz, Markus Albert, George Evangelou, André L.M. Oliveira, Ioana Gencia, Nada Vuckovic, Rosa Perelló, Ana Maria Draganita, Michel Colomb, Ayse Cefle, Hongguang Lu, Annarosa Virgili, Hayriye Saricaoglu, Esther A.W. Wolberink, Michael Russu, Elisabeth Arnoult-Coudoux, Caroline Nicaise-Bergère, Aleksandra M Ignjatović, Necmettin Özdemir, Kristīne Zabludovska, Cemal Bilaç, Jose Luis Lopez Estebaranz, Marie-Christine Lami, Harold S. Rabinovitz, Izabel Bota, Damien Grivet, Dimitrije Brasanac, Andrei Jalba, Joep Hoevenaars, Sofie De Schepper, Deniz Duman, Vladimir Vasku, Anna Belloni Fortina, Rosa Cristina Coppola, Marion Chavez-Bourgeois, Hoon-Soo Kim, Zamira Barragan, Julia Welzel, Thomas Ruzicka, Patricia V. Cristodor, Pierfrancesco Zampieri, Michael Lanthaler, Marc Haspeslagh, Jürgen Christian Becker, Gamze Erfan, Tanja Maier, Hui Mei Cheng, Mauro Enokihara, Ana Arance, Emel Dikicioglu Cetin, Pranaya A. Bagde, Mona M. Elfangary, Stefano Cavicchini, Alicia Barreiro, Odivânia Krüger, Mariana Petaccia Macedo, Itziar Erana Tomas, Elimar Elias Gomes, Monika Vrablova, Marcio Lorencini, Javier Alcántara González, Giuseppe Micali, Kerstin Kellermann, Mauricio Mendonca do Nascimento, Elisabeth Mt Wurm, Elena Sánchez-Largo Uceda, Yury Sergeev, Céleste Lebbé, Manfred Fiebiger, Gisele Gargantini Rezze, Antonio Graziano, Ana Pampín, Márcia Ferreira Candido, Martine Bagot, Jan Lapins, Nahide Onsun, Daniela Göppner, Katie Lee, Josef Schröder, Gisele G Rezze, Reyes Gamo, Mauricio Soto-Gamboa, Giovanni Pellacani, Maria Luiza P. Freitas, Mizuki Sawada, Hyun-Chang Ko, Ramon M Pujol Vallverdú, Jin gyoon Park, Peter Weber, Alberto Mota, Theofanis Spiliopoulos, Renata B. Marques, Daiji Furusho, Barbora Divisova, Pascale Guitera, Johan Heilborn, Alexandr Fedoseev, Athanasios Kyrgidis, Zakia Douhi, Mariame Meziane, Florent Grange, Alister Lilleyman, Juliana C. Marques-Da-Costa, Mitsuyasu Nakajima, Camilla Reggiani, Marina Meneses, Anna Sokolova, Zoe Apalla, Leo Čabrijan, Tim Lee, Piergiacomo Calzavara-Pinton, Tomas Fikrle, Georgios Chaidemenos, Braun Ralph, Aikaterini Patsatsi, Ekin Şavk, Marcela Pecora Cohen, Ioannis Efstratiou, Gurol Acikgoz, Pietro Quaglino, Nati Angelica, Luc Thomas, Edileia Bagatin, Kedima C. Nassif, Dimitrios Sotiriadis, Regina Fink-Puches, Anna Maria Wozniak, Salvador González, Agnieszka Buszko, Fezal Ozdemir, Banu Yaman, Vishnu Moodalgiri, Anne Grange, Robert J Meier, Davorin Loncaric, Fatmagül Keleş, Renato Marchiori Bakos, Sergio Chimenti, Sebastian Podlipnik, Pınar Incel Uysal, Devinder M Thappa, Nida Kaçar, Emel Bulbul Baskan, Erna Snellman, Pietro Rubegni, J. Kreusch, Hae Jin Pak, Danijela Dobrosavljevic Vukojevic, Bengü Nisa Akay, Holger A. Haenssle, Horacio Cabo, Anna Rammlmair, Fred Godtliebsen, Chiara Ferrari, Hiroshi Sakai, Christina Kemanetzi, Åsa Ingvar, Jitka Suchmannova, Zlata Janjic, Samira Zobiri, Haishan Zeng, Emine Böyük, Antonello Felli, Je-Ho Mun, Pablo Fernández Peñas, Ercan Caliskan, Satish S. Udare, Borna Pavičić, Max Hundeiker, Cristel Ruini, A. Hakan Cermik, Ülker Gül, Auro ra Parodi, Timothy P. Wu, Bernardo Gontijo, Ivan Klyuzhin, Gabriela Turcu, Sylvia Aidé Martínez-Cabriales, Francisco Alcántara Nicolás, Inge A. Krisanti, Sandra Cecilia García-García, Meriem Benfodda, Nika Madjlessi, Paraskevi Karagianni, Gizem Yağcıoğlu, Didem Dizman, Danielle I. Shitara, Nilda Eliana Gomez-Bernal, Mirna Šitum, Natalia Ilina, Job Van Der Heijden, Małgorzata Kwiatkowska, Bota Izabel, Ismini Vassilaki, Irene Potouridou, Jorge Luis Rosado, Lukas Prantl, María-José Bañuls, Fernando N. Barbosa, Seitaro Nakagawa, Jana Dornheim, Hitoshi Iyatomi, Rifat Saitburkhanov, Çiğdem Çağlayan, Natalie Ong, Stefano Gardini, Temeida Alendar, Zrinka Rendić-Miočević, Ryuhei Okuyama, Wafae Bono, Olga Warszawik-Hendzel, Danica Tiodorovic-Zivkovic, Alise Balcere, Ramazan Kahveci, Sebastian Gehmert, Herbert M. Kirchesch, Fernando Javier Pinedo, Raul Niin, Dan Savastru, Andreas Blum, Valeria Coco, Alexander C. Katoulis, Yosuke Yamamoto, Mumtaz Jabeen, Louise De Brot Andrade, Lidia Rudnicka, Pierre Wolkenstein, Fatma Pelin Cengiz, Woo-il Kim, Rainer Hofmann-Wellenhof, Tine Vestergaard, Maria Valeria B. Pinheiro, Ana Filipa Pedrosa, Caroline M. Takigami, Nilgün Bilen, Feroze Kaliyadan, Lotte Themstrup, Awatef Kelati, Katrien Vossaert, Burak Sezen, Natalia Jaimes, Olga Zhukova, Peter Jung, Nidhi Singh, Uxua Floristan, Ivette Alarcon, Michel Baccard, Flávia V. Bittencourt, Nicolas Dupin, Neslihan Şendur, Flavia Boff, Lydia Garcia Gaba, João Pedreira Duprat Neto, Caius Solovan, Byung Soo Kim, Anamaria Jović, Toshitsugu Sato, Antoni Bennassar, Ilkka Pölönen, Svetlana Rogozarski, Agnieszka Kardynał, Harald P.M. Gollnick, Anastasia Trigoni, Harvey Lui, Hiroshi Koga, Dai Ogata, Zeynep N. Saraçoğlu, Nilton B Rodrigues, Ketty Peris, Vanessa da Silva, Akira Hamada, Monica Corazza, Azmat A. Khan, Cengizhan Erdem, Victor Desmond Mandel, Sabina Zurac, Laura Elena Barbosa-Moreno, Filomena Azevedo, Matsue Hiroyuki, Philippe Saiag, Kara Shah, Stephen W. Dusza, Margaret Song, Francesca Giusti, Lidija Zolotarevski, Romain Vie, Rutao Cui, Aylin Okçu Heper, Kerstin Wöltje, Kyoko Tonomura, Charlotte H. Vuong, Moira Ragazzi, Marta Andreu Barasoain, Stephan Schreml, Branka Marinović, Mona R E Abdel Halim, Selimir Kovacevic, Noriaki Kamada, Adriana Garcia-Herrera, Ayse S. Filiz, Helena Collgros, Joan A. Puig-Butille, Ulvi Loite, Meng-Tsan Tsai, Nele Degryse, Philipp Tschandl, Seiichiro Wakabayashi, Korina Tzima, Kari Nielsen, Edith Arzberger, Alain Archimbaud, Makiko Miyamoto, Steffen Emmert, Katharine Hanlon, Stefano Astorino, Andre Sobiecki, Trevino A Pakasi, Giovanni Ghigliotti, Arzu Karataş Toğral, Sara Bassoli, Mahdi Akhbardeh, Martina Ulrich, Mirna Bradamante, Gökhan Uslu, Ross Flewell-Smith, Mauro Alaibac, Bettina Kranzelbinder, Steven Gazal, Nina Malishevskaya, Mikhail Ustinov, Noora Neittaanmäki-Perttu, Olga Simionescu, Saime Irkoren, Mahsa Ansari, Mustafa Turhan Sahin, Priit Kruus, Jana Janovska, Vesna Gajanin, Giovanni Ponti, Alon Scope, Ozkan Kanat, Cesare Massone, Thomas Schopf, Karolina Hadasik, Magnus Karlsson, Ayça Tan, Ignacio Gómez Martín, Armand Bensussan, Dilara Tüysüz, Saleh M. H. El Shiemy, Ine De Wispelaere, Malou Peppelman, Kenan Aydogan, Christian Teutsch, Ryszard A. Antkowiak, Nathalie De Carvahlo, Fatma Shabaka, Matthias Karasek, Christina Fotiadou, Wael M. Saudi, Matthias Weber, Maria Saletta Palumbo, Elisa Benati, Hana Helppikangas, Mariana Grigore, Leonard Witkamp, Rajiv Kumar, Stella Atkins, Eugene Y. Neretin, Dirk Berndt, Piet E.J van Erp, Alessandro Testori, David Duffy, Steluta Ratiu, Tara Bronsnick, Christoph Rinner, Soo-Han Woo, Federica Ferrari, Gabriela Garbin, Eduardo Nagore, Claus Duschl, Caterina Longo, Daniel Alcala-Perez, Helmut Beltraminelli, Sarah Hedtrich, David C McLean, Bojana Spasic, Martin Laimer, Malgorzata Pawlowska-Kisiel, Bohdan Lytvynenko, Heba I. Nagy Abd El-Gawad, Jean-Luc Perrot, Daška Štulhofer Buzina, Dimitrios Rigopoulos, Christian Hallermann, Jeffrey Keir, Adriana Martín Fuentes, Franz Trautinger, Walter L. G. Machado, Emese Gellén, Tatjana Ros, Gabriella Emri, Pinar Y. Basak, Nilay Duman, Reinhart Speeckaert, Peter Komericki, Maciel Zortea, Raphaela Kaestle, Lucía Pérez Carmona, Masaru Tanaka, Ionela Manole, Calin Giurcaneanu, Cristina Carrera, Jianhua Zhao, Marsha Mitchum, Isil Kilinc Karaarslan, Michael Muntifering, Alice Casari, Nicole Basset-Seguin, Seok-Kweon Yun, Vesna Mikulic, Albert Brugués, Kim-Dung Nguyen, Reshmi Madankumar, Joo-Ik Kim, Anna Skrok, Nicolle Mazzotti, Aomar Ammar-Khodja, Alina Avram, Laxmisha Chandrashekar, Dilek Biyik Ozkaya, Refika F. Artuz, Joanna Czuwara-Ladykowska, Hana Szakos, Dejan M Nikolic, Katarzyna Żórawicz, Georg Duftschmid, Natalia Pikelgaupt, Jorge Ocampo-Candiani, Irdina Drljevic, Canten Tataroglu, Esther Jiménez Blázquez, Philippe Gain, Simonetta Piana, Yunus Bulgu, Lars Dornheim, Bruno Labeille, Helmut Schaider, Nitul Khiroya, Sofia Theotokoglou, Christian Morsczeck, Kalliopi Armyra, Serap Öztürkcan, Shricharit h Shetty, Ozlem Su, Susana Puig, Lina Ivert, Katia Ongenae, Hirotsugu Shirabe, Ardalan Benam, Gustav Christensen, Veronika Paťavová, Adria Gual, Laura Pavoni, Mihaita Viorica Mihalceanu, Slobodan Jesic, Abdurrahman Bugra Cengiz, Jerome Becquart, Yasutomo Mikoshiba, Mattia Carbotti, Marcelo O. Samolé, Margherita Raucci, Sven Lanssens, Maria João M. Vasconcelos, Valeriy Semisazhenov, Fabio Facchetti, Monia Maccaferri, Vincenzo Panasiti, Camila M. Carvalho, Elena Tolomio, Ercan Arca, Celia Badenas, Sonia Segura Tigell, Francesco Lacarrubba, Ruzica Jurakic Toncic, Uday Khopkar, Uwe Seidl, Clóvis Antônio Lopes Pinto, Alice Marneffe, Zhenguo Wu, Josefin Lysell, Malgorzata Olszewska, Marta Ruano Del Salado, Alina Gogulescu, Tarl W. Prow, Christine Fink, Jean-Marie Tan, Milana Ivkov Simic, Mahshid S. Ansari, Stamatina Geleki, Sondang P. Sirait, Flavia Baderca, Marcella N. Silva, Andra Pehoiu, Joost Koehoorn, Ajay Goyal, Maria Dirlei Ferreira de Souza Begnami, Hui-bin Lu, Hoda A. Moneib, Maria Antonietta Pizzichetta, Scott Menzies, Gulsel Anil Bahali, Vesna Tlaker Zunter, Elfrida Carstea, Ines Chevolet, Septimiu Enache, Aysun Şikar Aktürk, Clara Kirchner, Greg Canning, Dina M. Shahin, Incilay Kalay Tugrul, Kristina Opletalova, Lars Hofmann, Mario Santinami, Anna Elisa Verzì, Asunción Vicente, Nathalia Delcourt, null Mernissi, Duru Tabanlıoglu Onan, Dorothy Polydorou, Irma Korom, Sara Moreno Fernández, Salim Gallouj, Annamari Ranki, Riina Hallik, Saduman Balaban Adim, Erietta Christofidou, Gustavo D. C. Dieamant, Vincenzo De Giorgi, Gregor B.E. Jemec, Kajsa Møllersen, Monisha lalji, Georgiana Simona Mohor, Hans-Jürgen Schulz, Justin R Sharpe, Karinna S. Machado, Efterpi Demiri, Mohammed I. AlJasser, Jelena Stojkovic-Filipovic, Harald Kittler, José M. A. Lopes, Adriana Diaconeasa, Patricia Serrano, Alfonso D’Orazio, Luca Mazzucchelli, Riccardo Bono, Oliver Felthaus, Juan Garcias-Ladaria, Zeljko Mijuskovic, Zsuzsanna Bago-Horvath, Alin Laurentiu Tatu, Christine Prodinger, Roland Blum, Demetrios Ioannides, Nadem Soufir, Diego Serraino, Ahmed M. Sadek, Leticia Calzado Villareal, Elliot Coates, Mariana Costache, Machuel Bruno, Bengu Gerceker Turk, Liliana Gabriela Popa, Han-Uk Kim, Lisa Hoogedoorn, Efstratios Vakirlis, Monika Kotrlá, Gabriel Salerni, Ela Comert, Salvatore Zanframundo, Zsuzsanna Lengyel, Francisco Jose Deleon, Maryam Sadeghi Naeeni, Georgios Kontochristopoulos, Ana Carolina Cherobin, Michiyo Matsumoto-Nakano, Gabriela Fortes Escobar, Maria Concetta Fargnoli, Ayse Oktem, Petra Fedorcova, Slavomir Urbancek, Hyunju Jin, Frédéric Cambazard, Tracey Newlove, Nataliya Sirmays, Cliff Rosendahl, Tamara Micantonio, Shirin Bajaj, Masa Gorsic, Ana Carolina L. Viana, Valentin Popa, Hubert Pehamberger, Anna Maria Carrozzo, Valentina Girgenti, Phil McClenahan, Beata Bergler-Czop, Alex Llambrich, Özgür Bakar, David Polsky, Krishnakant B. Pandya, Andrea Maurichi, Isabelle Hoorens, Paola Sorgi, Marianne Niin, Serena Magi, Malathi Munisamy, Zlatko Marušić, Cristina Mangas, Hakan Yesil, Miriam Potrony, Safaa Y. Negm, Maria T. Corradin, Stefania Seidenari, Işıl Bulur, Evelin Csernus, Gemma Tell-Marti, Alix Thomas, Juliana Casagrande Tavoloni Braga, Marco Manfredini, Karime M. Hassun, Celia Levy-Silbon, Lali Mekokishvili, Cem Yildirim, Hanna Eriksson, John H. Pyne, Angel Pizarro, Hakim Hammadi, Alessandro Borghi, Mariana A. Cordeiro, Fatima Zohra, A. Tülin Güleç, Ivan Ruiz Victoria, Joanna N. Łudzik, Radwa Magdy, Hisashi Uhara, Grażyna Kamińska-Winciorek, Llúcia Alòs, Pegah Kharazmi, Keisuke Suehiro, Lucian Russu, Zorica Đorđević Brlek, Sandrine Massart-Manil Massart-Manil, Moon-Bum Kim, Noha E. Hashem, Domenico Piccolo, Francesca Cicero, Jan Szymszal, Verena Ahlgrimm-Siess, Marian Gonzalez Inchaurraga, Ignazio Stanganelli, Danica Tiodorovic Zivkovic, Bugce Topukcu, Katharina Jaeger, Michael J. Inskip, Sara M. Mohy, Assya Djeridane, Véronique Del Marmol, Isil Kilinc, Nehal Yossif, Geon-Wook Kim, Oleksandr Litus, Ivana Ilić, Richard A Sturm, Mustafa Tunca, Anndressa da Matta, Elisabeth Jecel, Danijela Ćurković, Giuseppe Argenziano, Lynlee L. Lin, Elena Sotiriou, Mikela Petkovic, Suzana Kamberova, Sara Ibañes del Agua, Alan Cameron, Judit Oláh, Marc Nahuys, Leila Jeskanen, Zrinjka Paštar, Anna Wojas-Pelc, Ingela Ahnlide, Romana Čeović, Geoffrey Cains, Gilles Thuret, Mary Thomas, Marios Fragoulis, Drahomira Jarosikova, Manfred Beleut, Ferda Artüz, Brigitte Lavole, Francesco Todisco Grande, Carine Dal Pizzol, Erika Richtig, Nathalie Teixeira De Carvalho, Hans Peter Soyer, Amer M Alanazi, Vesna Sossi, Manal Bosseila, Monica Sulitan, Biancamaria Scoppio, Zrinka Bukvić Mokos, Marie-Jeanne P. Gerritsen, Mariano Suppa, Danielle Giambrone, Christoph Sinz, Jernej Kukovic, Martina Bosic, Adriana Rakowska, Eleni Mitsiou, Kely Hernandez, Ashfaq A. Marghoob, Daniel Boda, Alessandro Di Stefani, Luciana Trane, Leo Raudonikis, Akane Minagawa, Itaru Dekio, Athanassios Kyrgidis, Magdalena Wawrzynkiewicz, Katharina T Weiß, Chie Kamada, Lamberto Zara, Cristian Navarrete-Dechent, Serkan Yazici, Frédéric Renard, Leonie Mathemeier, Nissrine Amraoui, Mariana Fabris, Mariola Wyględowska-Kania, Nikolay Potekaev, Elisa Cinotti, Sedef Şahin, Peter van de Kerkhof, Silvana Ciardo, Sara Izzi, Paolo Piemonte, William V. Stoecker, Giampiero Mazzocchetti, Pasquale Frascione, Louise Lovatto, Ayşegül Yalçınkaya Iyidal, Jennifer A. Stein, Selçuk Yüksel, Daniela Ledić Drvar, Stine F. Pedersen, Dimitrios Sgouros, Meriem Bounouar, Balachandra S Ankad, Rahul Bute, Julia Brockley, Paula Aguilera-Otalvaro, Sumiko Ishizaki, Daniela Kulichova, Ilias Papadimitriou, Yeser Genc, Tanja Batinac, Jadran Bandic, Jean-Michel Lagarde, Göksun Karaman, Philipp Babilas, Mari Salmivuori, Lieven Annemans, Lennart K Blomqvist, Karel Pizinger, Duncan Lambie, Alexander Michael Witkowski, Meltem Uslu, Irena Savo, Martin Gosau, Raphaela Kastle, Olli Saksela, Pedro Zaballos, Esther De Eusebio Murillo, Hu Hui-Han, Sanda Mirela Cherciu, Claudia Artenie, Elvira Moscarella, Richard Johns, Ozlem Erdem, Valérie Vuong, Basma Birqdar, Jela Tomkova, Kasturee Jagirdar, Vassilios Lambropoulos, Moshira S. Bahrawy, Seong-Jin Kim, Su Chii Kong, Helen Schmid, Tetsuya Tsuchida, Michele Tonellato, Laura Berbegal, Lumír Pock, Iustin Hancu, Babar K Rao, Juliette Jegou, Lajos Kemény, Teresa Deinlein, Usha N. Khemani, Davive Guardoli, Juliana Arêas de Souza Lima Beltrame Ferreira, Tatiana Cristina Moraes Pinto Blumetti, Adhimukti T. Sampurna, Alexandru Telea, Ana Maria Forsea, Gionata Marazza, Lidija Kandolf Sekulovic, Marta Kurzeja, Marija Buljan, Fatima Zohra Mernissi, Alba Maiques-Diaz, Roger González, Dimitrios Kalabalikis, María Gabriela Vallone, Vanessa P. Martins Da Silva, Gemma Flores-Pons, Giuseppe Bertollo, Rolland Gyulai, Giuliana Crisman, Secil Saral, Simon Nicholson, Aimilios Lallas, Willeke Blokx, Marc A. L. M. Boone, and Oana Sindea
- Subjects
Oncology ,business.industry ,RL1-803 ,Genetics ,Medicine ,Library science ,Environmental ethics ,Dermatology ,business ,Molecular Biology - Published
- 2015
20. Development of 5-hydroxypyrazole derivatives as reversible inhibitors of lysine specific demethylase 1
- Author
-
Alison E. McGonagle, Daniel P. Mould, Tim C. P. Somervaille, Ulf Bremberg, Helen F. Small, Matthis Geitmann, Allan M. Jordan, Donald J. Ogilvie, and Alba Maiques-Diaz
- Subjects
0301 basic medicine ,animal structures ,Cancer therapy ,Reversible inhibitor ,Clinical Biochemistry ,Patent literature ,Pharmaceutical Science ,LSD1 ,Biochemistry ,Acute myeloid leukaemia ,Cell Line ,03 medical and health sciences ,Inhibitory Concentration 50 ,Mice ,Structure-Activity Relationship ,Catalytic Domain ,Drug Discovery ,Animals ,Humans ,Surface plasmon resonance ,Molecular Biology ,IC50 ,Histone Demethylases ,Binding Sites ,Manchester Cancer Research Centre ,Chemistry ,Epigenetic therapy ,ResearchInstitutes_Networks_Beacons/mcrc ,Organic Chemistry ,Rational design ,KDM1A ,Cell Differentiation ,Surface Plasmon Resonance ,Combinatorial chemistry ,Molecular Docking Simulation ,030104 developmental biology ,Stem cell differentiation ,Molecular Medicine ,Pyrazoles ,Epigenetics ,B7-2 Antigen ,LYSINE-SPECIFIC DEMETHYLASE 1 ,Half-Life - Abstract
A series of reversible inhibitors of lysine specific demethylase 1 (LSD1) with a 5-hydroxypyrazole scaffold have been developed from compound 7, which was identified from the patent literature. Surface plasmon resonance (SPR) and biochemical analysis showed it to be a reversible LSD1 inhibitor with an IC50 value of 0.23 µM. Optimisation of this compound by rational design afforded compounds with Kd values of
- Published
- 2017
21. The molecular pathogenesis of the NUP98-HOXA9 fusion protein in acute myeloid leukemia
- Author
-
Rocio N. Salgado, Mahesh Shrestha, Sara Alvarez, Alba Maiques-Diaz, Raúl Torres-Ruiz, Maria-Jose Calasanz, Ana Rio-Machin, Gonzalo Gómez-López, María José Larrayoz, Jude Fitzgibbon, F Garcia-Martinez, Juan C. Cigudosa, Claudia Haferlach, and Javier Munoz
- Subjects
0301 basic medicine ,LEUKEMOGENESIS ,Cancer Research ,Myeloid ,Oncogene Proteins, Fusion ,Transcription, Genetic ,Oncogene Proteins ,CD34(+) HEMATOPOIETIC-CELLS ,Biology ,THERAPY ,03 medical and health sciences ,Transcription (biology) ,medicine ,Humans ,Letter to the Editor ,Homeodomain Proteins ,PBX3 ,Gene Expression Profiling ,HEK 293 cells ,Myeloid leukemia ,NUP98/HOXA9 Fusion Protein ,Hematology ,HOXA9 ,medicine.disease ,Virology ,Nuclear Pore Complex Proteins ,Gene expression profiling ,MODEL ,Leukemia, Myeloid, Acute ,Leukemia ,HEK293 Cells ,030104 developmental biology ,medicine.anatomical_structure ,DIFFERENTIATION ,Oncology ,Cancer research - Abstract
Sí
- Published
- 2017
22. LSD1 inhibitors disrupt the GFI1 transcription repressor complex
- Author
-
Gary J. Spencer, Tim C. P. Somervaille, Alba Maiques-Diaz, and James T. Lynch
- Subjects
0301 basic medicine ,Cancer Research ,GFI1 ,medicine.medical_treatment ,LSD1 ,03 medical and health sciences ,0302 clinical medicine ,AML ,medicine ,Epigenetics ,Enhancer ,acetylation ,Manchester Cancer Research Centre ,epigenetics ,biology ,ResearchInstitutes_Networks_Beacons/mcrc ,Growth factor ,leukemia ,Myeloid leukemia ,KDM1A ,Methylation ,medicine.disease ,Cell biology ,Author's Views ,Leukemia ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Molecular Medicine ,Demethylase ,methylation - Abstract
Pharmacologic inhibition of KDM1A (Lysine Demethylase 1A) induces differentiation in certain subtypes of acute myeloid leukemia. Our recent studies reveal this is dependent upon drug-induced disruption of the GFI1 (Growth Factor Independent 1) transcription repressor complex, leading to activation of enhancers distributed close to genes controlling monocytic lineage differentiation.
- Published
- 2018
23. Downregulation of RUNX1/CBFβ by MLL fusion proteins enhances hematopoietic stem cell self-renewal
- Author
-
Stephen D. Nimer, Xiaomei Yan, Qianfei Wang, Bo Li, William Tse, Zhijian Xiao, Gang Huang, Yalan Rao, Rajeana M. Conway, Alba Maiques-Diaz, Wei Chen, James C. Mulloy, Nuomin Huang, Yue Zhang, Yunzhu Dong, Shannon Elf, Daniel G. Tenen, Xinghui Zhao, Yoshihiro Hayashi, Johannes Zuber, Aili Chen, and Fuhong He
- Subjects
Oncogene Proteins, Fusion ,Immunology ,Blotting, Western ,Transcription factor complex ,Biology ,Core binding factor ,Real-Time Polymerase Chain Reaction ,Biochemistry ,Core Binding Factor beta Subunit ,Translocation, Genetic ,chemistry.chemical_compound ,Mice ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,RNA, Messenger ,neoplasms ,Bone Marrow Transplantation ,Acute leukemia ,Myeloid Neoplasia ,Reverse Transcriptase Polymerase Chain Reaction ,Myeloid leukemia ,Cell Biology ,Hematology ,medicine.disease ,Flow Cytometry ,Hematopoietic Stem Cells ,Fusion protein ,Mice, Inbred C57BL ,Leukemia ,Leukemia, Myeloid, Acute ,Phenotype ,RUNX1 ,chemistry ,embryonic structures ,Core Binding Factor Alpha 2 Subunit ,Cancer research ,Myeloid-Lymphoid Leukemia Protein - Abstract
RUNX1/CBFβ (core binding factor [CBF]) is a heterodimeric transcription factor complex that is frequently involved in chromosomal translocations, point mutations, or deletions in acute leukemia. The mixed lineage leukemia (MLL) gene is also frequently involved in chromosomal translocations or partial tandem duplication in acute leukemia. The MLL protein interacts with RUNX1 and prevents RUNX1 from ubiquitin-mediated degradation. RUNX1/CBFβ recruits MLL to regulate downstream target genes. However, the functional consequence of MLL fusions on RUNX1/CBFβ activity has not been fully understood. In this report, we show that MLL fusion proteins and the N-terminal MLL portion of MLL fusions downregulate RUNX1 and CBFβ protein expression via the MLL CXXC domain and flanking regions. We confirmed this finding in Mll-Af9 knock-in mice and human M4/M5 acute myeloid leukemia (AML) cell lines, with or without MLL translocations, showing that MLL translocations cause a hypomorph phenotype of RUNX1/CBFβ. Overexpression of RUNX1 inhibits the development of AML in Mll-Af9 knock-in mice; conversely, further reducing Runx1/Cbfβ levels accelerates MLL-AF9–mediated AML in bone marrow transplantation assays. These data reveal a newly defined negative regulation of RUNX1/CBFβ by MLL fusion proteins and suggest that targeting RUNX1/CBFβ levels may be a potential therapy for MLLs.
- Published
- 2014
24. HDAC Inhibitors As Novel Targeted Therapies for NUP98-HOXA9 AML Patients
- Author
-
María José Calasanz, Sara Alvarez, Juan C. Cigudosa, Alba Maiques-Diaz, Gonzalo Gómez-López, Jude Fitzgibbon, and Ana Rio-Machin
- Subjects
Immunology ,Chromosomal translocation ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Fusion protein ,Chromatin ,homeobox A9 ,Downregulation and upregulation ,Regulatory sequence ,Cancer research ,Gene ,Transcription factor - Abstract
Background: The chromosomal translocation t(7;11)(p15,p15) encodes the oncogenic transcription factor NUP98-HOXA9 which results in a fusion of the nucleoporin 98kDa (NUP98) and homeobox A9 (HOXA9) genes. The oncogenic mechanisms underlying this translocation remain poorly understood and patients are currently inadequately served by traditional cytotoxic chemotherapy regimens. Aims:To decipher the underlying biology of the NUP98-HOXA9 fusion protein and develop rational therapeutic strategies targeting its oncogenic mechanism. Methods: Human cellular models expressing NUP98-HOXA9, HOXA9 wt or NUP98 wt were established by retroviral transduction of HEK293FT human cell line and human hematopoietic progenitors (CD34+, hHP) isolated from donor cord blood. Chromatin immunoprecepitation experiments followed by sequencing (ChIP-seq) and quantitative ChIP (qChIP) were used to define fusion specific binding locations. Cloning regulatory regions of selected target genes in a luciferase vectorconfirmed the direct involvement of NUP98-HOXA9 in their regulation. RTQ-PCR and gene expression microarrays were used to evaluate expression levels. Co-Immunoprecipitation experiments validated protein-protein interactions and drug treatments were performed at IC50. Cell viability was analysed by apoptosis, proliferation and Colony Forming Unit assays. Results:Comparison of ChIP-seq data from HEK293FTmodels of NUP98-HOXA9, HOXA9 wt or NUP98 wt respectively, identified 4,471 target genomic regions of the fusion protein (FDR < 0.05), located within +4/-4 kb from the annotated Transcription Start Site (TSS) of 1,363 genes, with 399 genes common to HOXA9 wt and 5 to NUP98 wt. The NUP98-HOXA9 binding sites included enhancers of MEIS1, HOXA9 and PBX3 (PBX3 and HOXA9 were common to NUP98 wt and MEIS1 to HOXA9 wt). Together these transcription factors form a key activator complex that regulates the expression of genes involved in leukemogenesis and its overexpression is significant related to adverse prognosis in AML. Luciferase assays showed that the upregulation of this leukemic axis was directly induced by the interaction of NUP98-HOXA9 with the corresponding enhancer regions of MEIS1, HOXA9 and PBX3. Treatment of cells with HXR9, a specific peptide inhibitor of HOXA9 and PBX3 interaction, led to a selective decrease in the proliferation of hHP expressing NUP98-HOXA9, confirming the relevance of these target genes to its oncogenic mechanism. Combining ChIP-seq and gene expression data of three independent clones of hHP expressing NUP98-HOXA9 and patient samples (n = 5) harbouring t(7;11)(p15,p15) revealed a dual regulatory role of the fusion protein, in both repressing and activating target gene transcription where, for example, MEIS1, HOXA9, PBX3 and AFF3 were found overexpressed and BIRC3, SMAD1, FILIP1L and PTEN downregulated. Interactions of NUP98-HOXA9 with p300 and HDAC1 were shown to drive this transcriptional activation and repression, respectively. We found using qChIP experiments that p300 bound to the regulatory regions of the overexpressed genes only when NUP98-HOXA9 was present, whereas we observed significant enrichment of HDAC1 binding to the promoter regions of the downregulated genes when the fusion protein was expressed. Taking advantage of this latter observation, we demonstrated a dramatic inhibitory effect on the viability of hHP expressing NUP98-HOXA9after the treatment with subtherapeutic doses (IC50 = 4nM) of the HDAC inhibitor LBH-589 (Panobinostat) with no effect in control hHP transduced with an empty vector. Conclusion: An improved understanding of the pathobiology underlying recurrent translocation events in AML is a critical first step for the development of rational, targeted therapies. Here, we identify upregulation of the targetable MEIS1-HOXA9-PBX3 complex underpinning the leukemogenic activity of NUP98-HOXA9. Its activity in repressing transcription mediated through interaction with HDAC1, has been shown to be also a key pathogenic mechanism that can be exploited through use of HDAC inhibitors and potentially lead to a promising new therapy for this high-risk group of patients. Disclosures No relevant conflicts of interest to declare.
- Published
- 2016
25. Pharmacological Inhibitors of LSD1 Promote Differentiation of Myeloid Leukemia Cells through a Mechanism Independent of Histone Demethylation
- Author
-
James T. Lynch, Filippo Ciceri, William J. Harris, Xu Huang, Tim C. P. Somervaille, Alba Maiques-Diaz, and Gary J. Spencer
- Subjects
animal structures ,Histone deacetylase 2 ,Cellular differentiation ,Immunology ,KDM1A ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Molecular biology ,Cell biology ,Histone H3 ,Histone demethylation ,Histone methylation ,Demethylase activity ,Histone demethylase activity - Abstract
Lysine Specific Demethylase 1 (LSD1 or KDM1A) is one of a number of epigenetic regulators which have recently emerged as candidate therapeutic targets in acute myeloid leukaemia (AML). It is a flavin adenine dinucleotide (FAD) dependent homolog of the amine oxidase family with an ability to demethylate monomethyl or dimethyl lysine 4 (K4) of histone H3, in addition to other substrates. Pharmacological inhibitors of LSD1 such as the tranylcypromine derivatives have already commenced evaluation in early phase clinical trials. While it has been widely assumed that these compounds promote differentiation of AML cells through inhibition of the demethylase activity of LSD1, the precise mechanisms by which LSD1 inhibitors function has not yet been determined. If changes in histone methylation are a central and critical mediator of the effects of LSD1 inhibitors in promoting AML cell differentiation, it would be expected that global changes in transcription would be tightly linked temporally to changes in histone methylation following drug treatment of cells. Through RNA sequencing and ChIP sequencing experiments performed in human THP1 AML cells treated for 24 hours with a potent and specific tranylcypromine-derivative LSD1 inhibitor (ORY86, trans-N-((2-methoxypyridin-3-yl)methyl)-2-phenylcyclopropan-1-amine), we have established that wholescale up regulation of a myeloid differentiation transcription programme occurs in the absence of any significant genome-wide changes in mono- and di-methyl H3K4 and H3K9 (which are key enzymatic targets of LSD1). Thus LSD1 inhibitor-induced up regulation of myeloid differentiation gene expression is not downstream of changes in histone methylation. We further demonstrated that non-enzymatic functions of LSD1 are essential in AML cells by expressing either wild-type (WT) or catalytically inactive LSD1 (K661A) in LSD1 knockdown (KD) THP1 cells. While LSD1 KD cells exhibit myeloid differentiation and loss of clonogenic potential, both the WT and mutant versions of LSD1 were able to rescue the in vitro clonogenic potential of KD cells to an equivalent extent. Thus the histone demethylase activity of LSD1 is not required to sustain AML blasts in an undifferentiated state. Comparison of the transcriptional consequences of LSD1 inhibition with the transcriptional consequences of transcription factor knockdown in THP1 AML cells using GSEA revealed that pharmacological inhibition of LSD1 mimics depletion of GFI1. Immunoprecipitation experiments confirmed the previously described physical association of GFI1 with LSD1. Critically, the physical interactions of LSD1 with GFI1 was reversed by pharmacological inhibition of LSD1 with ORY86. Furthermore, in ChIP sequencing experiments drug treatment led to dissociation of LSD1 from promoters and enhancers. By contrast, there was no disruption of the endogenous level interaction of LSD1 with RCOR1, HDAC1 and HDAC2 (i.e. the CoREST complex) following drug treatment. To determine whether the inhibitor-induced separation of LSD1 from GFI1 is required for induction of myeloid differentiation by ORY86, we performed experiments using a GFI1-LSD1 fusion construct expressed in THP1 cells under the control of a doxycycline-regulated promoter. This construct tethers LSD1 directly to the transcription factor and circumvents any drug induced physical separation. THP1 cells expressing GFI1-LSD1 were drug resistant (as determined by immunophenotyping and clonogenic potential), in contrast to control cells expressing GFI1, LSD1 or an empty vector in the same inducible system. Thus, drug-induced physical separation of GFI1 from LSD1 is required for THP1 AML cells to undergo differentiation. Our data support a model whereby the physical association of LSD1 with transcription factors such as GFI1 is essential to maintain the differentiation block in AML. Unexpectedly, tranylcypromine-derivative inhibitors target this novel scaffolding function of LSD1, rather than its histone demethylase activity, to promote differentiation of AML cells. Disclosures Lynch: Astra Zeneca: Employment. Ciceri:Oryzon Genomics: Employment. Somervaille:Oryzon Genomics: Research Funding; Imago Biosciences: Consultancy.
- Published
- 2014
26. RUNX1/CBFβ Dosage Is Critical for MLL Leukemias Development
- Author
-
William Tse, Bo Li, Yunzhu Dong, Qianfei Wang, Yue Zhang, Xinghui Zhao, Rajeana M. Conway, Gang Huang, Fuhong He, James C. Mulloy, Wei Chen, Stephen D. Nimer, Xiaomei Yan, Alba Maiques-Diaz, Zhijian Xiao, Shannon Elf, Daniel G. Tenen, Nuomin Huang, Johannes Zuber, Aili Chen, Yalan Rao, and Yoshihiro Hayashi
- Subjects
Acute leukemia ,Immunology ,Myeloid leukemia ,MLL Partial Tandem Duplication ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Fusion protein ,Molecular biology ,Haematopoiesis ,Leukemia ,chemistry.chemical_compound ,RUNX1 ,chemistry ,hemic and lymphatic diseases ,embryonic structures ,Cancer research ,medicine ,Progenitor cell ,neoplasms - Abstract
Transcription factors RUNX1/CBFβ play critical roles in hematopoiesis. Both of them are frequently involved in chromosomal translocations, point mutations, or deletions in acute leukemia. The mixed lineage leukemia (MLL) gene is also frequently involved in chromosomal translocations or partial tandem duplication in acute leukemia. We have previously shown that MLL, RUNX1, and CBFβ interact and form a regulatory complex to regulate downstream target genes. However, the functional consequence of MLL fusions on RUNX1/CBFβ activity remains unknown. To determine the impact of MLL fusion protein on RUNX1/CBFβ, we introduced either MLL, MLL-BP (longer N-terminal Flag-tagged MLL construct which contains CXXC domain; 1-1406), or MLL-fusions together with RUNX1, CBFβ, or both RUNX1 and CBFβ into 293T cells. MLL-BP and MLL fusions significantly decreased RUNX1 levels compared with controls (empty vector and MLL). CBFβ protein was mildly decreased by MLL-BP and MLL-fusions when expressed alone. However, when CBFβ was co-expressed with RUNX1, it was significantly decreased compared with controls. The expression levels of RUNX1 and CBFβ proteins in LSK cells from Mll-Af9 knock-in mice were significantly lower than those from wild-type (WT) mice. To confirm these findings in human acute myeloid leukemia (AML), we measured the expression of RUNX1 and CBFβ at both mRNA and protein levels in various leukemia cell lines. The expression levels of RUNX1 and CBFβ proteins were significantly decreased in AML cells with MLL fusion and MLL partial tandem duplication (MLL-PTD) compared with those in AML cells without MLL aberrations. MLL fusions still have CXXC domain. In MLL-PTD, the CXXC domain is duplicated. Our data showed that RUNX1 protein is not only down-regulated by MLL fusion proteins, but also by MLL-BP. Thus, to determine which region is involved in the down-regulation of RUNX1, we introduced a series of MLL deletion mutants into 293T cells and measured RUNX1 protein expression. MLL deletion mutants without CXXC domain had no effect on RUNX1 stability. The construct which contains point mutations in CXXC domain also lacked the ability to reduce RUNX1 expression. Furthermore, overexpression of only CXXC domain and flanking regions could down-regulate RUNX1 protein expression. These results suggest that MLL fusion proteins and the N-terminal MLL portion of MLL fusions down-regulate RUNX1 and CBFβ protein expression via the MLL CXXC domain and flanking regions. To understand the impact of RUNX1/CBFβ down-regulation on hematopoietic stem and progenitor cells (HSPCs), we generated RUNX1+/–/CBFβ+/– mice as a hypomorph model. The percentage of bone marrow (BM) LSK cells from RUNX1+/–/CBFβ+/– mice was significantly increased compared with that from WT mice. Using BM cells from these mice, we performed in vitro CFU assay and in vivo bone marrow transplantation (BMT) assay. BM cells from RUNX1+/–/CBFβ+/– mice provided more colonies in CFU assay compared with those from WT mice. To determine whether restoration of RUNX1 could repress the MLL mediated leukemogenesis, we retrovirally overexpressed WT RUNX1 in BM cells from Mll-Af9 knock-in mice. Using transduced BM cells, we performed in vitro CFU assay and in vivo BMT assay. RUNX1 overexpressed Mll-Af9 (Mll-Af9/RUNX1) cells underwent terminal differentiation after 2 times replating, while control vector transduced Mll-Af9 (Mll-Af9/Control) cells could still be replated more than 4 times. All the recipient mice transplanted with Mll-Af9/Control cells developed AML. In contrast, all the recipient mice transplanted with Mll-Af9/RUNX1 never develop AML. Furthermore, when we treated MLL leukemia cell lines with DOT1L inhibitor (EPZ-5676), RUNX1 protein levels in these MLL leukemia cell lines were significantly increased 48 hours after the treatment in comparing with controls treated with DMSO. However, there was no significant mRNA expression level change of RUNX1within 48 hours. Future studies are needed to fully understand the mechanism of whether this increasing RUNX1 protein level by DOT1L inhibitor is through blocking CXXC domain and flanking regions mediated degradation. In conclusion, MLL aberrations down-regulate RUNX1/CBFβ via their CXXC domain and flanking regions. Down-regulation of RUNX1/CBFβ plays critical role for MLL mediated leukemia development. Targeting RUNX1/CBFβ levels allows us to test novel therapies for MLL leukemias. Disclosures Mulloy: Celgene: Research Funding; Seattle Genetics: Research Funding; Amgen: Research Funding; NovImmune: Research Funding.
- Published
- 2014
27. Abstract 472: Interactions of the fusion protein Nup98-Hoxa9 with Pbx3, p300 and HDAC1: widening the targeted therapy window in acute myeloid leukemia (AML)
- Author
-
Raul M. Torres, Juan C. Cigudosa, Alba Maiques-Diaz, Sandra Rodriguez-Perales, Rocío Salgado, Juan C. Ramirez, Ana Rio-Machin, Álvaro Eguileor, and Sara Alvarez
- Subjects
Genetics ,Cancer Research ,Activator (genetics) ,Myeloid leukemia ,Biology ,medicine.disease ,Fusion protein ,Gene expression profiling ,DNA binding site ,Leukemia ,Oncology ,Cancer research ,medicine ,Gene ,Transcription factor - Abstract
The chromosomal translocation t(7;11)(p15,p15), that results in the oncogenic fusion protein Nup98-Hoxa9 (NH), appears in 1% of patients with AML and is associated with very poor prognosis and short overall survival. Despite the large severity of the leukemia induced by this fusion protein, the oncogenic events triggered by NH are poorly understood, although a potential role as an aberrant transcription factor has been proposed. We have generated a human Hematopoietic Progenitors (hHP) cellular model expressing NH constitutively to identify the molecular mechanisms supporting the malignancy of this fusion protein, facilitating the search for therapeutic targets. We identified the DNA binding sites of NH by performing ChIP-seq experiments, which were validated by qRT-PCR analysis on ChIP selected DNA and Luciferase assays. Expression profiling was performed in hHP-NH and co-Immunoprecipitations (Co-IPs) were done to demonstrate the interaction of NH with different transcriptional regulators. Specific drug sensitivity of the hHP-NH model was assessed in cell proliferation assays. Our work provides the first description of the DNA binding sites of NH, most of which are regulatory regions of genes involved in the development of AML. In particular, we demonstrate that NH induces the overexpression of MEIS1, HOXA9 and PBX3, transcription factors forming an activator complex that is a key element in the leukemic onset driven by other chromosome rearrangements. Interestingly, we show that NH directly interacts with this complex through Pbx3. To evaluate the biological relevance of the interaction of the MEIS1-HOXA9-PBX3 complex with NH, we have analyzed the sensitivity of hHP-NH to the HXR9 peptide (an inhibitor of the HOXA9-PBX3 interaction). Supporting our hypothesis, we observed an inhibitory effect on hHP-NH viability after HXR9 treatment. Finally, by combining the expression profile data from hHP-NH and the ChIP-seq results using GSEA analysis, we show that NH is able to induce both overexpression and down-regulation of its target genes. To provide evidences of the activator-repressor role of NH, we performed different Co-IPs that demonstrated its direct interaction with both p300 (transcriptional activator) and HDAC1 (transcriptional inhibitor). Taken together, we show that the direct overexpression of the complex MEIS1-HOXA9-PBX3 is one of the pathogenic mechanisms induced by NH. As expected, the disruption of this complex with the HXR9 peptide in the hHP-NH model has a direct effect on cell viability. Furthermore, we show that NH interacts with this complex via PBX3 and also with p300 and HDAC1. The features and architecture of these interactions need to be further explored, but these findings allow us to consider the use of the HXR9 peptide or some HDAC inhibitors as possible treatments for these patients. Citation Format: Ana Rio-Machin, Alba Maiques-Diaz, Sandra Rodriguez-Perales, Sara Alvarez, Rocio N. Salgado, Álvaro Eguileor, Raul Torres, Juan C. Ramirez, Juan C. Cigudosa. Interactions of the fusion protein Nup98-Hoxa9 with Pbx3, p300 and HDAC1: widening the targeted therapy window in acute myeloid leukemia (AML). [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 472. doi:10.1158/1538-7445.AM2014-472
- Published
- 2014
28. Chromatin Modifications Induced by the AML1/ETO Fusion Protein Reversibly Silence Its Genomic Targets Through AML1 and Sp1 Binding Motifs
- Author
-
Alba Maiques-Diaz, F V Jacinto, James C. Mulloy, María José Larrayoz, Fu-Sheng Chou, J C Cigudosa, Mark Wunderlich, Gonzalo Gómez-López, Sandra Rodriguez-Perales, Maria-Jose Calasanz, and Sara Alvarez
- Subjects
Chromatin Immunoprecipitation ,Cancer Research ,Oncogene Proteins, Fusion ,Sp1 Transcription Factor ,Immunology ,Histone Deacetylase 1 ,Real-Time Polymerase Chain Reaction ,Biochemistry ,Chromatin remodeling ,Epigenesis, Genetic ,Umbilical Cord ,Histones ,Histone H4 ,Fusion gene ,Histone H3 ,RUNX1 Translocation Partner 1 Protein ,Histone H1 ,hemic and lymphatic diseases ,Humans ,Histone code ,RNA, Messenger ,Promoter Regions, Genetic ,neoplasms ,Cells, Cultured ,Binding Sites ,biology ,Pioneer factor ,Acetylation ,Promoter ,Genomics ,Cell Biology ,Hematology ,Hematopoietic Stem Cells ,Molecular biology ,Chromatin ,ChIP-sequencing ,Leukemia, Myeloid, Acute ,Histone ,Oncology ,CTCF ,Core Binding Factor Alpha 2 Subunit ,biology.protein ,Histone deacetylase ,Chromatin immunoprecipitation - Abstract
Abstract 2422 The AML1/ETO fusion protein, which is present in 10% to 15% of cases of acute myeloid leukemia (AML), is known to repress myeloid differentiation genes through DNA binding and recruitment of chromatin-modifying proteins and transcription factors in target genes. Previous studies have shown that both histone H4 deacetylation and K9 trimethylation of histone H3 (H3K9me3), respectively catalyzed by enzymes such as histone deacetylase 1 (HDAC1) and lysine methyltransferase enzyme suppressor of variegation 3–9 homolog 1(SUV39H1), are direct AML1/ETO effects on specific locus, but to date no global analysis of the AML1/ETO functional repressive chromatin modifications have been performed. ChIP-chip analysis of human hematopoietic stem/progenitor cells transduced with the AML1/ETO fusion gene (HSPC-AE) enabled us to identify 1168 AML1/ETO target genes, 103 of which were co-occupied by HDAC1 with a lost of hyperacetylation at histone H4, and 264 showed an increase H3K9me3 (p(X)0.7). We found a significant enrichment of genes involved in hematopoietic differentiation and in specific signaling pathways (i.e. TGF-β and Wnt/β-catenin) in AML1/ETO epigenetically modified target gene sets. Among them we identified novel genes (i.e. CTCF, MAPK1, SIRT1, YES1, AML1, MLLT3, and RPS19) previously described to be involved on the hematopoietic development, which epigenetic repression could implicate relevant steps in AML development induced by t(8;21). Gene Set Enrichment Analysis (GSEA) showed that expression of the 103 AML1/ETO-HDAC1 and 264 AML1/ETO-H3K9me3 target genes is highly diminished on HSCP-AE cells compared to the normal HSPC (p Disclosures: No relevant conflicts of interest to declare.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.