1
|
Koyama S, Liu X, Koike Y, Hikino K, Koido M, Li W, Akaki K, Tomizuka K, Ito S, Otomo N, Suetsugu H, Yoshino S, Akiyama M, Saito K, Ishikawa Y, Benner C, Natarajan P, Ellinor PT, Mushiroda T, Horikoshi M, Ikeda M, Iwata N, Matsuda K, Niida S, Ozaki K, Momozawa Y, Ikegawa S, Takeuchi O, Ito K, Terao C. Population-specific putative causal variants shape quantitative traits. Nat Genet 2024; 56:2027-2035. [PMID: 39363016 PMCID: PMC11525193 DOI: 10.1038/s41588-024-01913-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/14/2024] [Indexed: 10/05/2024]
Abstract
Human genetic variants are associated with many traits through largely unknown mechanisms. Here, combining approximately 260,000 Japanese study participants, a Japanese-specific genotype reference panel and statistical fine-mapping, we identified 4,423 significant loci across 63 quantitative traits, among which 601 were new, and 9,406 putatively causal variants. New associations included Japanese-specific coding, splicing and noncoding variants, exemplified by a damaging missense variant rs730881101 in TNNT2 associated with lower heart function and increased risk for heart failure (P = 1.4 × 10-15 and odds ratio = 4.5, 95% confidence interval = 3.1-6.5). Putative causal noncoding variants were supported by state-of-art in silico functional assays and had comparable effect sizes to coding variants. A plausible example of new mechanisms of causal variants is an enrichment of causal variants in 3' untranslated regions (UTRs), including the Japanese-specific rs13306436 in IL6 associated with pro-inflammatory traits and protection against tuberculosis. We experimentally showed that transcripts with rs13306436 are resistant to mRNA degradation by regnase-1, an RNA-binding protein. Our study provides a list of fine-mapped causal variants to be tested for functionality and underscores the importance of sequencing, genotyping and association efforts in diverse populations.
Collapse
Affiliation(s)
- Satoshi Koyama
- Laboratory for Cardiovascular Genomics and Informatics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Xiaoxi Liu
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yoshinao Koike
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory for Bone and Joint Diseases, RIKEN Center for Medical Sciences, Tokyo, Japan
- Department of Orthopedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Keiko Hikino
- Laboratory for Pharmacogenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Masaru Koido
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Cancer Biology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Wei Li
- Department of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kotaro Akaki
- Department of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kohei Tomizuka
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Shuji Ito
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory for Bone and Joint Diseases, RIKEN Center for Medical Sciences, Tokyo, Japan
- Department of Orthopedic Surgery, Shimane University Faculty of Medicine, Izumo, Japan
| | - Nao Otomo
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory for Bone and Joint Diseases, RIKEN Center for Medical Sciences, Tokyo, Japan
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hiroyuki Suetsugu
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory for Bone and Joint Diseases, RIKEN Center for Medical Sciences, Tokyo, Japan
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Soichiro Yoshino
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masato Akiyama
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Ocular Pathology and Imaging Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kohei Saito
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
| | - Yuki Ishikawa
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Christian Benner
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Pradeep Natarajan
- Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Personalized Medicine, Mass General Brigham, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Patrick T Ellinor
- Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Taisei Mushiroda
- Laboratory for Pharmacogenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Momoko Horikoshi
- Laboratory for Genomics of Diabetes and Metabolism, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Masashi Ikeda
- Department of Psychiatry, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Nakao Iwata
- Department of Psychiatry, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Koichi Matsuda
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Shumpei Niida
- Research Institute, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kouichi Ozaki
- Laboratory for Cardiovascular Genomics and Informatics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Research Institute, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Shiro Ikegawa
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory for Bone and Joint Diseases, RIKEN Center for Medical Sciences, Tokyo, Japan
| | - Osamu Takeuchi
- Department of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kaoru Ito
- Laboratory for Cardiovascular Genomics and Informatics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan.
- Department of Applied Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan.
| |
Collapse
|
2
|
Liu N, Kawahira N, Nakashima Y, Nakano H, Iwase A, Uchijima Y, Wang M, Wu SM, Minamisawa S, Kurihara H, Nakano A. Notch and retinoic acid signals regulate macrophage formation from endocardium downstream of Nkx2-5. Nat Commun 2023; 14:5398. [PMID: 37669937 PMCID: PMC10480477 DOI: 10.1038/s41467-023-41039-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 08/15/2023] [Indexed: 09/07/2023] Open
Abstract
Hematopoietic progenitors are enriched in the endocardial cushion and contribute, in a Nkx2-5-dependent manner, to tissue macrophages required for the remodeling of cardiac valves and septa. However, little is known about the molecular mechanism of endocardial-hematopoietic transition. In the current study, we identified the regulatory network of endocardial hematopoiesis. Signal network analysis from scRNA-seq datasets revealed that genes in Notch and retinoic acid (RA) signaling are significantly downregulated in Nkx2-5-null endocardial cells. In vivo and ex vivo analyses validate that the Nkx2-5-Notch axis is essential for the generation of both hemogenic and cushion endocardial cells, and the suppression of RA signaling via Dhrs3 expression plays important roles in further differentiation into macrophages. Genetic ablation study revealed that these macrophages are essential in cardiac valve remodeling. In summary, the study demonstrates that the Nkx2-5/Notch/RA signaling plays a pivotal role in macrophage differentiation from hematopoietic progenitors.
Collapse
Affiliation(s)
- Norika Liu
- The Jikei University School of Medicine, Department of Cell Physiology, Tokyo, Japan
- University of California Los Angeles, Department of Molecular Cell and Developmental Biology, Los Angeles, USA
| | - Naofumi Kawahira
- University of California Los Angeles, Department of Molecular Cell and Developmental Biology, Los Angeles, USA
| | | | - Haruko Nakano
- University of California Los Angeles, Department of Molecular Cell and Developmental Biology, Los Angeles, USA
| | - Akiyasu Iwase
- University of Tokyo, Department of Physiological Chemistry and Metabolism, Tokyo, Japan
| | - Yasunobu Uchijima
- University of Tokyo, Department of Physiological Chemistry and Metabolism, Tokyo, Japan
| | - Mei Wang
- The Jikei University School of Medicine, Department of Cell Physiology, Tokyo, Japan
| | - Sean M Wu
- Stanford University, Cardiovascular Institute and Division of Cardiovascular Medicine, Department of Medicine, Stanford, USA
| | - Susumu Minamisawa
- The Jikei University School of Medicine, Department of Cell Physiology, Tokyo, Japan
| | - Hiroki Kurihara
- University of Tokyo, Department of Physiological Chemistry and Metabolism, Tokyo, Japan
| | - Atsushi Nakano
- The Jikei University School of Medicine, Department of Cell Physiology, Tokyo, Japan.
- University of California Los Angeles, Department of Molecular Cell and Developmental Biology, Los Angeles, USA.
- University of California Los Angeles, David Geffen Department of Medicine, Division of Cardiology, Los Angeles, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, USA.
| |
Collapse
|
3
|
Azevedo Portilho N, Scarfò R, Bertesago E, Ismailoglu I, Kyba M, Kobayashi M, Ditadi A, Yoshimoto M. B1 lymphocytes develop independently of Notch signaling during mouse embryonic development. Development 2021; 148:271231. [PMID: 34370006 PMCID: PMC8380455 DOI: 10.1242/dev.199373] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 07/08/2021] [Indexed: 11/20/2022]
Abstract
B1 lymphocytes are a small but unique component of the innate immune-like cells. However, their ontogenic origin is still a matter of debate. Although it is widely accepted that B1 cells originate early in fetal life, whether or not they arise from hematopoietic stem cells (HSCs) is still unclear. In order to shed light on the B1 cell origin, we set out to determine whether their lineage specification is dependent on Notch signaling, which is essential for the HSC generation and, therefore, all derivatives lineages. Using mouse embryonic stem cells (mESCs) to recapitulate murine embryonic development, we have studied the requirement for Notch signaling during the earliest B-cell lymphopoiesis and found that Rbpj-deficient mESCs are able to generate B1 cells. Their Notch independence was confirmed in ex vivo experiments using Rbpj-deficient embryos. In addition, we found that upregulation of Notch signaling induced the emergence of B2 lymphoid cells. Taken together, these findings indicate that control of Notch signaling dose is crucial for different B-cell lineage specification from endothelial cells and provides pivotal information for their in vitro generation from PSCs for therapeutic applications. This article has an associated ‘The people behind the papers’ interview. Summary: During development, the first B lymphocytes are produced in the absence of Notch signaling and, thus, independently from HSCs.
Collapse
Affiliation(s)
- Nathalia Azevedo Portilho
- Center for Stem Cell Research and Regenerative Medicine, Institute for Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Rebecca Scarfò
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Elisa Bertesago
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Ismail Ismailoglu
- Lillehei Heart Institute and Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA.,Genetics and Development Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Michael Kyba
- Lillehei Heart Institute and Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michihiro Kobayashi
- Center for Stem Cell Research and Regenerative Medicine, Institute for Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Andrea Ditadi
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Momoko Yoshimoto
- Center for Stem Cell Research and Regenerative Medicine, Institute for Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
4
|
A systems biology pipeline identifies regulatory networks for stem cell engineering. Nat Biotechnol 2019; 37:810-818. [PMID: 31267104 DOI: 10.1038/s41587-019-0159-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 05/16/2019] [Indexed: 12/18/2022]
Abstract
A major challenge for stem cell engineering is achieving a holistic understanding of the molecular networks and biological processes governing cell differentiation. To address this challenge, we describe a computational approach that combines gene expression analysis, previous knowledge from proteomic pathway informatics and cell signaling models to delineate key transitional states of differentiating cells at high resolution. Our network models connect sparse gene signatures with corresponding, yet disparate, biological processes to uncover molecular mechanisms governing cell fate transitions. This approach builds on our earlier CellNet and recent trajectory-defining algorithms, as illustrated by our analysis of hematopoietic specification along the erythroid lineage, which reveals a role for the EGF receptor family member, ErbB4, as an important mediator of blood development. We experimentally validate this prediction and perturb the pathway to improve erythroid maturation from human pluripotent stem cells. These results exploit an integrative systems perspective to identify new regulatory processes and nodes useful in cell engineering.
Collapse
|
5
|
Bigas A, Porcheri C. Notch and Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:235-263. [DOI: 10.1007/978-3-319-89512-3_12] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
6
|
Li X, Liu F, Zhang X, Shi G, Ren J, Ji J, Ding L, Fan H, Dou H, Hou Y. Notch-Hes-1 axis controls TLR7-mediated autophagic death of macrophage via induction of P62 in mice with lupus. Cell Death Dis 2016; 7:e2341. [PMID: 27537524 PMCID: PMC5108329 DOI: 10.1038/cddis.2016.244] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 07/11/2016] [Accepted: 07/12/2016] [Indexed: 01/21/2023]
Abstract
The increased death of macrophages has been considered as a pathogenic factor for systemic lupus erythematosus (SLE), and dysfunction of autophagy may contribute to improper cell death. However, the effect of autophagy on macrophage during the pathogenesis of SLE is still unclear. Here we found that the death rate and autophagy level of macrophages significantly increased in MRL/lpr lupus-prone mice. Activation of toll-like receptor 7 (TLR7) triggered macrophage death in an autophagy-dependent but caspase-independent way in vitro. Moreover, P62/SQSTM1 is thought to have an essential role in selective autophagy. We also demonstrated that P62/SQSTM1 was required for TLR7-induced autophagy, and knockdown of P62 suppressed R848-induced cell death and LC3II protein accumulation. As an important mediator for cell-cell communication, Notch signaling is responsible for cell-fate decisions. Our results showed that activation of TLR7 also upregulated the expression of Notch1, especially its downstream target gene Hairy and enhancer of split 1 (Hes-1) in macrophages. Of note, we found that Hes-1, as a transcriptional factor, controlled TLR7-induced autophagy by regulating P62 expression. Furthermore, to confirm the above results in vivo, TLR7 agonist imiquimod (IMQ)-induced lupus mouse model was prepared. Splenic macrophages from IMQ-treated mice exhibited increased autophagy and cell death as well as enhanced expressions of Notch1 and Hes-1. Our results indicate that Notch1-Hes-1 signaling controls TLR7-induced autophagic death of macrophage via regulation of P62 in mice with lupus.
Collapse
Affiliation(s)
- Xiaojing Li
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Fei Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Xuefang Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Guoping Shi
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Jing Ren
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Jianjian Ji
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Liang Ding
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Hongye Fan
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Huan Dou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| |
Collapse
|
7
|
García-Alegría E, Lafita-Navarro MC, Aguado R, García-Gutiérrez L, Sarnataro K, Ruiz-Herguido C, Martín F, Bigas A, Canelles M, León J. NUMB inactivation confers resistance to imatinib in chronic myeloid leukemia cells. Cancer Lett 2016; 375:92-99. [PMID: 26944313 DOI: 10.1016/j.canlet.2016.02.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Revised: 02/23/2016] [Accepted: 02/23/2016] [Indexed: 01/21/2023]
Abstract
Chronic myeloid leukemia (CML) progresses from a chronic to a blastic phase, where the leukemic cells are proliferative and undifferentiated. The CML is nowadays successfully treated with BCR-ABL kinase inhibitors as imatinib and its derivatives. NUMB is an evolutionary well-conserved protein initially described as a functional antagonist of NOTCH function. NUMB is an endocytic protein associated with receptor internalization, involved in multiple cellular functions. It has been reported that MSI2 protein, a NUMB inhibitor, is upregulated in CML blast crisis, whereas NUMB itself is downregulated. This suggest that NUMB plays a role in the malignant progression of CML. Here we have generated K562 cells (derived from CML in blast crisis) constitutively expressing a dominant negative form of NUMB (dnNUMB). We show that dnNUMB expression confers a high proliferative phenotype to the cells. Importantly, dnNUMB triggers a partial resistance to imatinib in these cells, antagonizing the apoptosis mediated by the drug. Interestingly, imatinib resistance is not linked to p53 status or NOTCH signaling, as K562 lack p53 and imatinib resistance is reproduced in the presence of NOTCH inhibitors. Taken together, our data support the hypothesis that NUMB activation could be a new therapeutic target in CML.
Collapse
Affiliation(s)
- Eva García-Alegría
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria and Dpto. de Biología Molecular, Universidad de Cantabria, Santander, Spain
| | - M Carmen Lafita-Navarro
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria and Dpto. de Biología Molecular, Universidad de Cantabria, Santander, Spain
| | - Rocío Aguado
- Instituto de Parasitología y Biomedicina, CSIC, P. T. Ciencias de la Salud, Granada, Spain
| | - Lucia García-Gutiérrez
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria and Dpto. de Biología Molecular, Universidad de Cantabria, Santander, Spain
| | - Kyle Sarnataro
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria and Dpto. de Biología Molecular, Universidad de Cantabria, Santander, Spain
| | | | | | - Anna Bigas
- Stem Cells and Cancer Group. IMIM, Barcelona, Spain
| | - Matilde Canelles
- Instituto de Parasitología y Biomedicina, CSIC, P. T. Ciencias de la Salud, Granada, Spain.
| | - Javier León
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria and Dpto. de Biología Molecular, Universidad de Cantabria, Santander, Spain.
| |
Collapse
|
8
|
Butko E, Pouget C, Traver D. Complex regulation of HSC emergence by the Notch signaling pathway. Dev Biol 2015; 409:129-138. [PMID: 26586199 DOI: 10.1016/j.ydbio.2015.11.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 11/11/2015] [Accepted: 11/12/2015] [Indexed: 01/13/2023]
Abstract
Hematopoietic stem cells are formed during embryonic development, and serve as the foundation of the definitive blood program for life. Notch signaling has been well established as an essential direct contributor to HSC specification. However, several recent studies have indicated that the contribution of Notch signaling is complex. HSC specification requires multiple Notch signaling inputs, some received directly by hematopoietic precursors, and others that occur indirectly within neighboring somites. Of note, proinflammatory signals provided by primitive myeloid cells are needed for HSC specification via upregulation of the Notch pathway in hemogenic endothelium. In addition to multiple requirements for Notch activation, recent studies indicate that Notch signaling must subsequently be repressed to permit HSC emergence. Finally, Notch must then be reactivated to maintain HSC fate. In this review, we discuss the growing understanding of the dynamic contributions of Notch signaling to the establishment of hematopoiesis during development.
Collapse
Affiliation(s)
- Emerald Butko
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Claire Pouget
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - David Traver
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA.
| |
Collapse
|
9
|
Influence of Jagged1 on apoptosis-related gene expression: a microarray database analysis. Genes Genomics 2015. [DOI: 10.1007/s13258-015-0316-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
10
|
Notch1 acts via Foxc2 to promote definitive hematopoiesis via effects on hemogenic endothelium. Blood 2015; 125:1418-26. [PMID: 25587036 DOI: 10.1182/blood-2014-04-568170] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Hematopoietic and vascular development share many common features, including cell surface markers and sites of origin. Recent lineage-tracing studies have established that definitive hematopoietic stem and progenitor cells arise from vascular endothelial-cadherin(+) hemogenic endothelial cells of the aorta-gonad-mesonephros region, but the genetic programs underlying the specification of hemogenic endothelial cells remain poorly defined. Here, we discovered that Notch induction enhances hematopoietic potential and promotes the specification of hemogenic endothelium in differentiating cultures of mouse embryonic stem cells, and we identified Foxc2 as a highly upregulated transcript in the hemogenic endothelial population. Studies in zebrafish and mouse embryos revealed that Foxc2 and its orthologs are required for the proper development of definitive hematopoiesis and function downstream of Notch signaling in the hemogenic endothelium. These data establish a pathway linking Notch signaling to Foxc2 in hemogenic endothelial cells to promote definitive hematopoiesis.
Collapse
|
11
|
Cell interactions and cell signaling during hematopoietic development. Exp Cell Res 2014; 329:200-6. [DOI: 10.1016/j.yexcr.2014.10.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 10/02/2014] [Accepted: 10/05/2014] [Indexed: 12/30/2022]
|
12
|
Guiu J, Bergen DJM, De Pater E, Islam ABMMK, Ayllón V, Gama-Norton L, Ruiz-Herguido C, González J, López-Bigas N, Menendez P, Dzierzak E, Espinosa L, Bigas A. Identification of Cdca7 as a novel Notch transcriptional target involved in hematopoietic stem cell emergence. ACTA ACUST UNITED AC 2014; 211:2411-23. [PMID: 25385755 PMCID: PMC4235648 DOI: 10.1084/jem.20131857] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Guiu et al. use ChIP-on-chip analysis for the Notch partner RBPj, using embryonic tissue from the aorta-gonad-mesonephros region to identify potential novel Notch target genes involved in HSC emergence. They show that c-MYC–responsive gene Cdca7 is expressed in different HSC and progenitor subpopulations and that CDCA7 is important for maintaining the undifferentiated phenotype. Cdca7 acts downstream of Notch in HSCs in zebrafish, mouse, and human, indicating a highly conserved Notch/RBPj/Cdca7 axis in hematopoietic development. Hematopoietic stem cell (HSC) specification occurs in the embryonic aorta and requires Notch activation; however, most of the Notch-regulated elements controlling de novo HSC generation are still unknown. Here, we identify putative direct Notch targets in the aorta-gonad-mesonephros (AGM) embryonic tissue by chromatin precipitation using antibodies against the Notch partner RBPj. By ChIP-on-chip analysis of the precipitated DNA, we identified 701 promoter regions that were candidates to be regulated by Notch in the AGM. One of the most enriched regions corresponded to the Cdca7 gene, which was subsequently confirmed to recruit the RBPj factor but also Notch1 in AGM cells. We found that during embryonic hematopoietic development, expression of Cdca7 is restricted to the hematopoietic clusters of the aorta, and it is strongly up-regulated in the hemogenic population during human embryonic stem cell hematopoietic differentiation in a Notch-dependent manner. Down-regulation of Cdca7 mRNA in cultured AGM cells significantly induces hematopoietic differentiation and loss of the progenitor population. Finally, using loss-of-function experiments in zebrafish, we demonstrate that CDCA7 contributes to HSC emergence in vivo during embryonic development. Thus, our study identifies Cdca7 as an evolutionary conserved Notch target involved in HSC emergence.
Collapse
Affiliation(s)
- Jordi Guiu
- Program de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Parc de Recerca Biomèdica de Barcelona, 08003 Barcelona, Spain
| | - Dylan J M Bergen
- Program de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Parc de Recerca Biomèdica de Barcelona, 08003 Barcelona, Spain
| | - Emma De Pater
- Erasmus MC Stem Cell and Regenerative Medicine Institute, Erasmus Medical Center, 3000 CA Rotterdam, Netherlands
| | - Abul B M M K Islam
- Research Unit on Biomedical Informatics, Department of Experimental and Health Sciences, Pompeu Fabra University, 08003 Barcelona, Spain Department of Genetic Engineering and Biotechnology, University of Dhaka, Dhaka 1000, Bangladesh
| | - Verónica Ayllón
- Centre for Genomics and Oncological Research (Genyo), Pfizer-University of Granada-Andalusian Government, 18016 Granada, Spain
| | - Leonor Gama-Norton
- Program de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Parc de Recerca Biomèdica de Barcelona, 08003 Barcelona, Spain
| | - Cristina Ruiz-Herguido
- Program de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Parc de Recerca Biomèdica de Barcelona, 08003 Barcelona, Spain
| | - Jessica González
- Program de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Parc de Recerca Biomèdica de Barcelona, 08003 Barcelona, Spain
| | - Nuria López-Bigas
- Research Unit on Biomedical Informatics, Department of Experimental and Health Sciences, Pompeu Fabra University, 08003 Barcelona, Spain Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Pablo Menendez
- José Carreras Leukaemia Research Institute, Cell Therapy Program, School of Medicine, University of Barcelona, 08036 Barcelona, Spain Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Elaine Dzierzak
- Erasmus MC Stem Cell and Regenerative Medicine Institute, Erasmus Medical Center, 3000 CA Rotterdam, Netherlands
| | - Lluis Espinosa
- Program de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Parc de Recerca Biomèdica de Barcelona, 08003 Barcelona, Spain
| | - Anna Bigas
- Program de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Parc de Recerca Biomèdica de Barcelona, 08003 Barcelona, Spain
| |
Collapse
|
13
|
Cortegano I, Melgar-Rojas P, Luna-Zurita L, Siguero-Álvarez M, Marcos MAR, Gaspar ML, de la Pompa JL. Notch1 regulates progenitor cell proliferation and differentiation during mouse yolk sac hematopoiesis. Cell Death Differ 2014; 21:1081-94. [PMID: 24583642 DOI: 10.1038/cdd.2014.27] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 12/27/2013] [Accepted: 01/28/2014] [Indexed: 01/08/2023] Open
Abstract
Loss-of-function studies have demonstrated the essential role of Notch in definitive embryonic mouse hematopoiesis. We report here the consequences of Notch gain-of-function in mouse embryo hematopoiesis, achieved by constitutive expression of Notch1 intracellular domain (N1ICD) in angiopoietin receptor tyrosine kinase receptor-2 (Tie2)-derived enhanced green fluorescence protein (EGFP(+)) hematovascular progenitors. At E9.5, N1ICD expression led to the absence of the dorsal aorta hematopoietic clusters and of definitive hematopoiesis. The EGFP(+) transient multipotent progenitors, purified from E9.5 to 10.5 Tie2-Cre;N1ICD yolk sac (YS) cells, had strongly reduced hematopoietic potential, whereas they had increased numbers of hemogenic endothelial cells. Late erythroid cell differentiation stages and mature myeloid cells (Gr1(+), MPO(+)) were also strongly decreased. In contrast, EGFP(+) erythro-myeloid progenitors, immature and intermediate differentiation stages of YS erythroid and myeloid cell lineages, were expanded. Tie2-Cre;N1ICD YS had reduced numbers of CD41(++) megakaryocytes, and these produced reduced below-normal numbers of immature colonies in vitro and their terminal differentiation was blocked. Cells from Tie2-Cre;N1ICD YS had a higher proliferation rate and lower apoptosis than wild-type (WT) YS cells. Quantitative gene expression analysis of FACS-purified EGFP(+) YS progenitors revealed upregulation of Notch1-related genes and alterations in genes involved in hematopoietic differentiation. These results represent the first in vivo evidence of a role for Notch signaling in YS transient definitive hematopoiesis. Our results show that constitutive Notch1 activation in Tie2(+) cells hampers YS hematopoiesis of E9.5 embryos and demonstrate that Notch signaling regulates this process by balancing the proliferation and differentiation dynamics of lineage-restricted intermediate progenitors.
Collapse
Affiliation(s)
- I Cortegano
- 1] Immunology Department, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Ctra. Majadahonda-Pozuelo, km 2, 28220 Madrid, Spain [2] Centro de Biología Molecular, Consejo Superior de Investigaciones Científicas (CBM-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain
| | - P Melgar-Rojas
- Program of Cardiovascular Developmental Biology, Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - L Luna-Zurita
- Program of Cardiovascular Developmental Biology, Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - M Siguero-Álvarez
- Program of Cardiovascular Developmental Biology, Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - M A R Marcos
- Centro de Biología Molecular, Consejo Superior de Investigaciones Científicas (CBM-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain
| | - M L Gaspar
- Immunology Department, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Ctra. Majadahonda-Pozuelo, km 2, 28220 Madrid, Spain
| | - J L de la Pompa
- Program of Cardiovascular Developmental Biology, Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| |
Collapse
|
14
|
Fraser ST. The modern primitives: applying new technological approaches to explore the biology of the earliest red blood cells. ISRN HEMATOLOGY 2013; 2013:568928. [PMID: 24222861 PMCID: PMC3814094 DOI: 10.1155/2013/568928] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 08/25/2013] [Indexed: 01/01/2023]
Abstract
One of the most critical stages in mammalian embryogenesis is the independent production of the embryo's own circulating, functional red blood cells. Correspondingly, erythrocytes are the first cell type to become functionally mature during embryogenesis. Failure to achieve this invariably leads to in utero lethality. The recent application of technologies such as transcriptome analysis, flow cytometry, mutant embryo analysis, and transgenic fluorescent gene expression reporter systems has shed new light on the distinct erythroid lineages that arise early in development. Here, I will describe the similarities and differences between the distinct erythroid populations that must form for the embryo to survive. While much of the focus of this review will be the poorly understood primitive erythroid lineage, a discussion of other erythroid and hematopoietic lineages, as well as the cell types making up the different niches that give rise to these lineages, is essential for presenting an appropriate developmental context of these cells.
Collapse
Affiliation(s)
- Stuart T. Fraser
- Disciplines of Physiology, Anatomy and Histology, Bosch Institute, School of Medical Sciences, University of Sydney, Medical Foundation Building K25, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
| |
Collapse
|
15
|
Roy A, Haldar S, Basak NP, Banerjee S. Molecular cross talk between Notch1, Shh and Akt pathways during erythroid differentiation of K562 and HEL cell lines. Exp Cell Res 2013; 320:69-78. [PMID: 24095799 DOI: 10.1016/j.yexcr.2013.09.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 09/04/2013] [Accepted: 09/26/2013] [Indexed: 11/20/2022]
Abstract
Erythropoiesis is a tightly regulated process dependent on extrinsic signals conveyed by the bone marrow niche. The signalling pathways thus activated or repressed do not act in isolation; rather an intricate cross talk among these pathways ensues homoeostasis within the erythroid compartment. In this study, we describe the effects of two such signalling pathways namely the Notch1 and the Shh pathway on erythropoiesis in immortalised K562 and HEL cell lines as well as the cross talk that ensues between them. We show that while activation of the Notch1 pathway inhibits differentiation of erythroid lineage cell lines as well as in in-vitro primary erythroid cultures from the human CD34(+) cells; Shh pathway favours erythroid differentiation. Further, the Notch1 pathway activates the Akt pathway and constitutively active Akt partially mimics the effect of Notch1 activation on erythropoiesis. Moreover, the Notch1, Akt and Shh pathways were found to cross talk with each other. In this process, activation of Notch1 was found to down regulate the Shh pathway independent of Akt activation. Significantly, Notch1 not only down regulated the Shh pathway, but also inhibited recombinant Shh mediated erythropoiesis. Our study thus reveals an intricate crosstalk among the Notch1, Shh and Akt pathways wherein Notch1 emerges as a key regulator of erythropoiesis.
Collapse
Affiliation(s)
- Anita Roy
- Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata-700064, India
| | | | | | | |
Collapse
|
16
|
Huang C, Jackson M, Samuel K, Taylor AH, Lowell S, Forrester LM. Haematopoietic differentiation is inhibited when Notch activity is enhanced in FLK1(+) mesoderm progenitors. Stem Cell Res 2013; 11:1273-87. [PMID: 24064354 DOI: 10.1016/j.scr.2013.08.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 08/08/2013] [Accepted: 08/26/2013] [Indexed: 12/16/2022] Open
Abstract
Notch signalling has been implicated during haematopoietic development in vivo and in the differentiation of haematopoietic cells from pluripotent cells in vitro. However interpretation of data from many of these studies has been complicated by the heterogeneous nature of cell populations under study and by the fact that the Notch pathway is active during embryogenesis prior to the development of the haematopoietic system. To define the role of Notch signalling in more precise cell populations during the early stages of haematopoietic development within the aorta-gonad-mesonephros (AGM) microenvironment we co-cultured differentiating ESCs on a stromal cell line derived from this region of the embryo. Our co-culture system had no effect on the production of FLK1(+) mesoderm progenitor cells but promoted their subsequent haematopoietic differentiation. We assessed the role of Notch signalling on haematopoietic differentiation of isolated FLK1(+) cells. Notch activity is dynamic and drops to basal levels as FLK1(+) cells commit to a haematopoietic fate. Further reduction of Notch activity by the inducible expression of dominant negative MAML had no functional consequences. In contrast, induction of Notch activity using an inducible NotchIC expression system had an inhibitory effect on haematopoietic differentiation. We used a Cre-mediated recombination strategy whereby NotchIC-expressing cells were marked with the hCD2 receptor and observed a reduction in the number of multi-lineage and myeloid colonies derived from NotchIC(+) compared to NotchIC(-) FLK1(+) cells isolated from the same culture. We believe that our culture system represents a good model for haematopoietic development within the AGM microenvironment and our data suggest that haematopoietic commitment of FLK1(+) cells in this setting occurs when Notch activity is below a specific threshold.
Collapse
Affiliation(s)
- Caoxin Huang
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | | | | | | | | | | |
Collapse
|
17
|
Guiu J, Shimizu R, D'Altri T, Fraser ST, Hatakeyama J, Bresnick EH, Kageyama R, Dzierzak E, Yamamoto M, Espinosa L, Bigas A. Hes repressors are essential regulators of hematopoietic stem cell development downstream of Notch signaling. ACTA ACUST UNITED AC 2012; 210:71-84. [PMID: 23267012 PMCID: PMC3549704 DOI: 10.1084/jem.20120993] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Previous studies have identified Notch as a key regulator of hematopoietic stem cell (HSC) development, but the underlying downstream mechanisms remain unknown. The Notch target Hes1 is widely expressed in the aortic endothelium and hematopoietic clusters, though Hes1-deficient mice show no overt hematopoietic abnormalities. We now demonstrate that Hes is required for the development of HSC in the mouse embryo, a function previously undetected as the result of functional compensation by de novo expression of Hes5 in the aorta/gonad/mesonephros (AGM) region of Hes1 mutants. Analysis of embryos deficient for Hes1 and Hes5 reveals an intact arterial program with overproduction of nonfunctional hematopoietic precursors and total absence of HSC activity. These alterations were associated with increased expression of the hematopoietic regulators Runx1, c-myb, and the previously identified Notch target Gata2. By analyzing the Gata2 locus, we have identified functional RBPJ-binding sites, which mutation results in loss of Gata2 reporter expression in transgenic embryos, and functional Hes-binding sites, which mutation leads to specific Gata2 up-regulation in the hematopoietic precursors. Together, our findings show that Notch activation in the AGM triggers Gata2 and Hes1 transcription, and next HES-1 protein represses Gata2, creating an incoherent feed-forward loop required to restrict Gata2 expression in the emerging HSCs.
Collapse
Affiliation(s)
- Jordi Guiu
- Program in Cancer Research, Hospital del Mar Medical Research Institute, IMIM, Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
GATA-1 utilizes Ikaros and polycomb repressive complex 2 to suppress Hes1 and to promote erythropoiesis. Mol Cell Biol 2012; 32:3624-38. [PMID: 22778136 DOI: 10.1128/mcb.00163-12] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The transcription factor Hairy Enhancer of Split 1 (HES1), a downstream effector of the Notch signaling pathway, is an important regulator of hematopoiesis. Here, we demonstrate that in primary erythroid cells, Hes1 gene expression is transiently repressed around proerythroblast stage of differentiation. Using mouse erythroleukemia cells, we found that the RNA interference (RNAi)-mediated depletion of HES1 enhances erythroid cell differentiation, suggesting that this protein opposes terminal erythroid differentiation. This is also supported by the decreased primary erythroid cell differentiation upon HES1 upregulation in Ikaros-deficient mice. A comprehensive analysis led us to determine that Ikaros favors Hes1 repression in erythroid cells by facilitating recruitment of the master regulator of erythropoiesis GATA-1 alongside FOG-1, which mediates Hes1 repression. GATA-1 is then necessary for the chromatin binding of the NuRD remodeling complex ATPase MI-2, the transcription factor GFI1B, and the histone H3K27 methyltransferase EZH2 along with Polycomb repressive complex 2. We show that EZH2 is required for the transient repression of Hes1 in erythroid cells. In aggregate, our results describe a mechanism whereby GATA-1 utilizes Ikaros and Polycomb repressive complex 2 to promote Hes1 repression as an important step in erythroid cell differentiation.
Collapse
|
19
|
Abstract
Notch is a well-conserved signaling pathway and its function in cell fate determination is crucial in embryonic development and in the maintenance of tissue homeostasis during adult life. Notch activation depends on cell-cell interactions that are essential for the generation of cell diversity from initially equivalent cell populations. In the adult hematopoiesis, Notch is undoubtedly a very efficient promoter of T-cell differentiation, and this has masked for a long time the effects of Notch on other blood lineages, which are gradually being identified. However, the adult hematopoietic stem cell (HSC) remains mostly refractory to Notch intervention in experimental systems. In contrast, Notch is essential for the generation of the HSCs, which takes place during embryonic development. This review summarizes the knowledge accumulated in recent years regarding the role of the Notch pathway in the different stages of HSC ontology from embryonic life to fetal and adult bone marrow stem cells. In addition, we briefly examine other systems where Notch regulates specific stem cell capacities, in an attempt to understand how Notch functions in stem cell biology.
Collapse
|
20
|
Bennani-Baiti B, Bennani-Baiti IM. Gene symbol precision. Gene 2012; 491:103-9. [DOI: 10.1016/j.gene.2011.09.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 09/21/2011] [Accepted: 09/29/2011] [Indexed: 11/26/2022]
|
21
|
Abstract
Hematopoiesis is the process that generates all the cell types of the blood, which are responsible for oxygen transport and immune defense. It has been now more than 50 years from the demonstration that blood cells derive from a common ancestor called Hematopoietic Stem Cell (HSC) McCulloch and Till (1960). Thus, the hematopoietic process relies on the unlimited and distinctive self-renewal ability of HSC, which in the adult mammalian organisms reside in the bone marrow, but their generation occurs during embryonic life. Questions still remain about how HSCs acquire and maintain the features of self-renewal and pluripotency that define stem-cell populations. Notch is a crucial signaling pathway involved in the generation of cell diversity and stem-cell maintenance in different systems. In some cases, Notch prevents differentiation, while in other contexts Notch directly participates in promoting cell differentiation. In the following sections, we will review what is known about the role of Notch in HSC establishment and hematopoietic cell lineage specification.
Collapse
|
22
|
Abstract
Notch is a crucial cell signaling pathway in metazoan development. By means of cell-cell interactions, Notch signaling regulates cellular identity, proliferation, differentiation and apoptosis. Within the last decade, numerous studies have shown an important role for this pathway in the development and homeostasis of mammalian stem cell populations. Hematopoietic stem cells (HSCs) constitute a well-defined population that shows self-renewal and multi-lineage differentiation potential, with the clinically relevant capacity to repopulate the hematopoietic system of an adult organism. Here, we review the emergence, development and maintenance of HSCs during mammalian embryogenesis and adulthood, with respect to the role of Notch signaling in hematopoietic biology.
Collapse
|
23
|
Deletion of Adam10 in endothelial cells leads to defects in organ-specific vascular structures. Blood 2011; 118:1163-74. [PMID: 21652679 DOI: 10.1182/blood-2011-04-348557] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
During vertebrate angiogenesis, Notch regulates the cell-fate decision between vascular tip cells versus stalk cells. Canonical Notch signaling depends on sequential proteolytic events, whereby interaction of Notch with membrane-anchored ligands triggers proteolytic processing, first by Adam10 and then presenilins. This liberates the Notch intracellular domain, allowing it to enter the nucleus and activate Notch-dependent genes. Here we report that conditional inactivation of Adam10 in endothelial cells (A10ΔEC) recapitulates the increased branching and density of the retinal vasculature that is also caused by interfering with Notch signaling. Moreover, A10ΔEC mice have additional vascular abnormalities, including aberrant subcapsular hepatic veins, enlarged glomeruli, intestinal polyps containing endothelial cell masses, abnormal endochondral ossification, leading to stunted long bone growth and increased pathologic neovascularization following oxygen-induced retinopathy. Our findings support a model in which Adam10 is a crucial regulator of endothelial cell-fate decisions, most likely because of its essential role in canonical Notch signaling.
Collapse
|
24
|
Sun Y, Gao X, Liu J, Kong QY, Wang XW, Chen XY, Wang Q, Cheng YF, Qu XX, Li H. Differential Notch1 and Notch2 expression and frequent activation of Notch signaling in gastric cancers. Arch Pathol Lab Med 2011; 135:451-8. [PMID: 21466361 DOI: 10.5858/2009-0665-oa.1] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT The biologic effects of Notch1 and Notch2 vary with cancer types and their potential role(s) in gastric cancers (GCs) remains largely unknown. OBJECTIVES This study aimed to address the previously mentioned issue by checking the expression of Notch1, Notch2, and Notch target gene Hes1 in GCs, premalignant gastric lesions, and noncancerous endoscopic gastric mucosa and by inhibiting Notch signal transduction in GC cells. DESIGN The status of Notch1, Notch2, and Hes1 expression in 74 GC surgical specimens, 10 endoscopic samples, and 4 human GC cell lines was evaluated by tissue microarray-based immunohistochemical staining, Western blotting, and reverse transcription-polymerase chain reaction, and the importance of Notch signaling was elucidated by treating 2 GC cell lines with 2 γ-secretase inhibitors. RESULTS Notch1 was undetectable in noncancerous gastric mucosa but was expressed with nuclear translocation in 16.7% (4 of 24) of chronic gastritis, 50.0% (9 of 18) of intestinal metaplasia, 54.2% (26 of 48) of intestinal GC, and 23.1% (6 of 26) of diffuse GC, showing distinct differences of Notch1 detection rates between either intestinal metaplasia and chronic gastritis or intestinal GCs and diffuse GCs (P = .03; P = .005, respectively). Notch2 nuclear translocation frequencies were 10.0% (1 of 10) in noncancerous endoscopic mucosa, 71.4% (30 of 42) in premalignant lesions, and 97.3% (72 of 74) in GC tissues, demonstrating a correlation of Notch2 expression with both intestinal GC and diffuse GC formation (P < .001). The rates of nuclear-Hes1 labeling were 1 of 10 among noncancerous, 42.9% premalignant, and 81.1% cancer tissues, which were closely correlated with Notch2 (P < .001) rather than Notch1 (P = .42) nuclear translocation. Only Notch2 was expressed accompanied with Hes1 nuclear labeling in the 4 GC cell lines established from diffuse GC cases. Inhibition of Notch signaling with γ-secretase inhibitors, L-685,458 and DAPT, prevented Hes1 nuclear translocation but neither suppressed growth nor induced cell death. CONCLUSIONS This study demonstrated a close correlation of Notch2 expression with GC formation and the potential link of Notch1 upregulation with intestinal-like phenotypes of gastric lesions. Although inhibition of Notch activity failed to achieve anti-GC effects, the activated Notch signaling may reflect a potential GC risk.
Collapse
Affiliation(s)
- Yuan Sun
- Liaoning Laboratory of Cancer Genomics and Department of Cell Biology, Dalian Medical University, Dalian, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
The Notch2-Jagged1 interaction mediates stem cell factor signaling in erythropoiesis. Cell Death Differ 2010; 18:371-80. [PMID: 20829885 DOI: 10.1038/cdd.2010.110] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Stem cell factor (SCF), the ligand for the c-kit receptor, is essential for the production of red blood cells during development and stress erythropoiesis. SCF promotes erythroblast proliferation and survival, while delaying erythroid differentiation through mechanisms that are largely unknown. In cultures of primary human differentiating erythroblasts, we found that SCF induces an increase in the expression of Notch2, a member of the Notch family implicated in the control of cell growth and differentiation. Functional inhibition of either Notch or its ligand Jagged1 inhibited the effects of SCF on erythroid cell expansion. SCF also induced the expression of Hes-1 and GATA-2, which may contribute to transduce Notch2 signals in response to SCF. Transduction of primary erythroid precursors with a dominant-negative Notch2 mutant inhibited both basal and SCF-mediated erythroblast expansion, and counteracted the effects of SCF on erythroblast differentiation. These findings provide a clue to understand the effects of increased proliferation and delayed differentiation elicited by SCF on the erythroid compartment and indicate Notch2 as a new player in the regulation of red cell differentiation.
Collapse
|
26
|
Gering M, Patient R. Notch signalling and haematopoietic stem cell formation during embryogenesis. J Cell Physiol 2010; 222:11-6. [PMID: 19725072 DOI: 10.1002/jcp.21905] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The Notch signalling pathway is repeatedly employed during embryonic development and adult homeostasis of a variety of tissues. In particular, its frequent involvement in the regulation of stem and progenitor cell maintenance and proliferation, as well as its role in binary fate decisions in cells that are destined to differentiate, is remarkable. Here, we review its role in the development of haematopoietic stem cells during vertebrate embryogenesis and put it into the context of Notch's functions in arterial specification, angiogenic vessel sprouting and vessel maintenance. We further discuss interactions with other signalling cascades, and pinpoint open questions and some of the challenges that lie ahead.
Collapse
Affiliation(s)
- Martin Gering
- Institute of Genetics, School of Biology, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | | |
Collapse
|
27
|
Nutlin-3 up-regulates the expression of Notch1 in both myeloid and lymphoid leukemic cells, as part of a negative feedback antiapoptotic mechanism. Blood 2009; 113:4300-8. [DOI: 10.1182/blood-2008-11-187708] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Abstract
The small molecule inhibitor of the MDM2/p53 interaction Nutlin-3 significantly up-regulated the steady-state mRNA and protein levels of Notch1 in TP53wild-type (OCI, SKW6.4) but not in TP53deleted (HL-60) or TP53mutated (BJAB) leukemic cell lines. A direct demonstration that NOTCH1 was a transcriptional target of p53 in leukemic cells was obtained in experiments carried out with siRNA for p53. Moreover, inhibition of Notch1 expression using Notch1-specific siRNA significantly increased cytotoxicity in TP53wild-type leukemic cells. Of note, Nutlin-3 up-regulated Notch1 expression also in primary TP53wild-type B-chronic lymphocytic leukemia (B-CLL) cells and the combined use of Nutlin-3 plus pharmacological γ-secretase inhibitors of the Notch signaling showed a synergistic cytotoxicity in both TP53wild-type leukemic cell lines and primary B-CLL cells. A potential drawback of γ-secretase inhibitors was their ability to enhance osteoclastic maturation of normal circulating preosteoclasts induced by RANKL + M-CSF. Notwithstanding, Nutlin-3 completely suppressed osteoclastogenesis irrespective of the presence of γ-secretase inhibitors. Taken together, these data indicate that the p53-dependent up-regulation of Notch1 in response to Nutlin-3 represents an antiapoptotic feedback mechanism able to restrain the potential therapeutic efficacy of Nutlin-3 in hematologic malignancies. Therefore, therapeutic combinations of Nutlin-3 + γ-secretase inhibitors might potentiate the cytotoxicity of Nutlin-3 in p53wild-type leukemic cells.
Collapse
|
28
|
Peller S, Tabach Y, Rotschild M, Garach-Joshua O, Cohen Y, Goldfinger N, Rotter V. Identification of gene networks associated with erythroid differentiation. Blood Cells Mol Dis 2009; 43:74-80. [PMID: 19329339 DOI: 10.1016/j.bcmd.2009.01.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Accepted: 01/20/2009] [Indexed: 10/21/2022]
Abstract
Erythropoiesis is a multistep process involving a large number of genes, which balance between proliferation, differentiation and survival of the erythroid cells. To understand the molecular mechanisms of erythropoiesis and related pathological aberrations, we analyzed three stages of in vitro differentiating human erythroid cells by expression profiling. We identified distinct clusters of genes, each with a unique expression pattern during differentiation. As JAK2 was shown to play a central role in myeloproliferative disorders, we focused on one cluster which includes JAK2 and other genes with high correlation to JAK2 expression. These genes had a low expression at the early erythroblast which increased in the intermediate stage and further slightly increased in the last stage of differentiation. Our results indicate that gene networks may associate with JAK2 expression in erythroid differentiation. It is intriguing to determine whether the pathogenesis of polycythemia vera (PV), harboring a common or uncommon JAK2 mutation, involves alterations in independent gene pathways that underlie the normal erythropoietic process.
Collapse
Affiliation(s)
- Shoshana Peller
- The Laboratory of Hematology, Assaf-Harofeh Medical Center, Zerifin, Israel.
| | | | | | | | | | | | | |
Collapse
|
29
|
Qiao L, Li GHY, Dai Y, Wang J, Li Z, Zou B, Gu Q, Ma J, Pang R, Lan HY, Wong BCY. Gene expression profile in colon cancer cells with respect to XIAP expression status. Int J Colorectal Dis 2009; 24:245-60. [PMID: 18704457 DOI: 10.1007/s00384-008-0566-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/29/2008] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIMS We observed a marked synergism between peroxisome proliferator-activated receptor gamma (PPARgamma) ligands and X-linked inhibitor of apoptosis (XIAP) down-regulation in colon cancer. In the current study, we detected the gene expression profile in HCT116 cells treated with or without PPARgamma ligand troglitazone. MATERIALS AND METHODS HCT116-XIAP(+/+) and HCT116-XIAP(-/-) cells were treated with or without 50 microM troglitazone for 48 h. Gene expressions were detected by microarray, and selected genes were validated by reverse-transcriptase polymerase chain reaction (PCR), real-time PCR, and Western blot. RESULTS Relative to HCT116-XIAP(+/+) cells, 58 genes were up-regulated and 33 genes down-regulated in HCT116-XIAP(-/-) cells, all by > or =4-fold. These genes could be classified into a wide variety of functional classes, but we focused on those related to angiogenesis, apoptosis, and proliferation. Thus, two pro-apoptotic genes and one pro-proliferation gene were up-regulated in HCT116-XIAP(-/-) cells. Two pro-proliferation genes, one pro-angiogenesis gene, one anti-angiogenesis gene, and one anti-apoptosis gene were down-regulated in HCT116-XIAP(-/-) cells. Relative to HCT116-XIAP(+/+) cells treated with troglitazone, 137 genes were up-regulated, and 31 genes were down-regulated in troglitazone-treated HCT116-XIAP(-/-) cells, all by > or =4-fold. Among the up-regulated genes were two anti-angiogenesis genes, seven pro-apoptosis genes, and six anti-proliferation genes. Among the down-regulated genes were one anti-angiogenesis gene, one pro-angiogenesis gene, one anti-apoptosis gene, one anti-proliferation gene, and two pro-proliferation genes. CONCLUSION Down-regulation of XIAP in HCT116 cells with or without troglitazone treatment was associated with changes of gene expression that favor increased tendency of apoptosis, decreased cell proliferation, and angiogenesis potential.
Collapse
Affiliation(s)
- Liang Qiao
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Krejcí A, Bernard F, Housden BE, Collins S, Bray SJ. Direct response to Notch activation: signaling crosstalk and incoherent logic. Sci Signal 2009; 2:ra1. [PMID: 19176515 DOI: 10.1126/scisignal.2000140] [Citation(s) in RCA: 135] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Notch is the receptor in one of a small group of conserved signaling pathways that are essential at multiple stages in development. Although the mechanism of transduction impinges directly on the nucleus to regulate transcription through the CSL [CBF-1/Su(H)/LAG-1] [corrected] DNA binding protein, there are few known direct target genes. Thus, relatively little is known about the immediate cellular consequences of Notch activation. We therefore set out to determine the genome-wide response to Notch activation by analyzing the changes in messenger RNA (mRNA) expression and the sites of CSL occupancy within 30 minutes of activating Notch in Drosophila cells. Through combining these data, we identify high-confidence direct targets of Notch that are implicated in the maintenance of adult muscle progenitors in vivo. These targets are enriched in cell morphogenesis genes and in components of other cell signaling pathways, especially the epidermal growth factor receptor (EGFR) pathway. Also evident are examples of incoherent network logic, where Notch stimulates the expression of both a gene and the repressor of that gene, which may result in a transient window of competence after Notch activation. Furthermore, because targets comprise both positive and negative regulators, cells become poised for both outcomes, suggesting one mechanism through which Notch activation can lead to opposite effects in different contexts.
Collapse
Affiliation(s)
- Alena Krejcí
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | | | | | | | | |
Collapse
|
31
|
Cheng X, Huber TL, Chen VC, Gadue P, Keller GM. Numb mediates the interaction between Wnt and Notch to modulate primitive erythropoietic specification from the hemangioblast. Development 2008; 135:3447-58. [PMID: 18799543 PMCID: PMC3039875 DOI: 10.1242/dev.025916] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
During embryonic development, the establishment of the primitive erythroid lineage in the yolk sac is a temporally and spatially restricted program that defines the onset of hematopoiesis. In this report, we have used the embryonic stem cell differentiation system to investigate the regulation of primitive erythroid development at the level of the hemangioblast. We show that the combination of Wnt signaling with inhibition of the Notch pathway is required for the development of this lineage. Inhibition of Notch signaling at this stage appears to be mediated by the transient expression of Numb in the hemangioblast-derived blast cell colonies. Activation of the Notch pathway was found to inhibit primitive erythropoiesis efficiently through the upregulation of inhibitors of the Wnt pathway. Together, these findings demonstrate that specification of the primitive erythroid lineage is controlled, in part, by the coordinated interaction of the Wnt and Notch pathways, and position Numb as a key mediator of this process.
Collapse
Affiliation(s)
- Xin Cheng
- Department of Gene and Cell Medicine; Mount Sinai School of Medicine; New York, NY 10029, USA
| | - Tara L. Huber
- Stem Cell and Developmental Biology Department; Genome Institute of Singapore 138672; Singapore
| | - Vincent C. Chen
- Department of Gene and Cell Medicine; Mount Sinai School of Medicine; New York, NY 10029, USA
| | - Paul Gadue
- Department of Gene and Cell Medicine; Mount Sinai School of Medicine; New York, NY 10029, USA
| | - Gordon M. Keller
- McEwen Centre for Regenerative Medicine; University Health Network; Toronto, Ontario, Canada
| |
Collapse
|
32
|
Chadwick N, Fennessy C, Nostro MC, Baron M, Brady G, Buckle AM. Notch induces cell cycle arrest and apoptosis in human erythroleukaemic TF-1 cells. Blood Cells Mol Dis 2008; 41:270-7. [PMID: 18693120 DOI: 10.1016/j.bcmd.2008.06.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Revised: 05/13/2008] [Accepted: 06/17/2008] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Notch signalling is known to promote hematopoietic stem cell self-renewal and to influence the lineage commitment decisions of progenitor cells. The purpose of this study was to investigate the mechanism of Notch-induced apoptosis in the erythroleukaemic cell line TF-1, and in primary cord blood CD34+ cells. METHODS Retroviral constructs containing constitutively active forms of Notch as well as components of the Notch signalling pathway were used to transduce cells and their effect on cell cycle kinetics and apoptosis assayed by immunostaining for the S-phase marker Ki67 and Annexin V. RESULTS We found that TF-1 cells undergo cell cycle arrest followed by apoptosis in a cytokine-independent manner in response to active Notch. Transduction of TF-1 cells with known targets of Notch signalling, Deltex1, HES1 and HERP2, showed that Notch-induced cell cycle arrest was not mediated by these proteins. However, analysis of cell cycle gene expression revealed that Notch signalling was associated with an up-regulation of IFI16 expression in TF-1 cells and in primary cord blood CD34+ cells. CONCLUSION These data demonstrate that, in the context of TF-1 cells, Notch signalling can induce cell cycle arrest and apoptosis.
Collapse
Affiliation(s)
- Nicholas Chadwick
- Faculty of Life Sciences, Manchester Interdisciplinary Biocenter, University of Manchester, Manchester M1 7DN, UK
| | | | | | | | | | | |
Collapse
|
33
|
Robert-Moreno À, Guiu J, Ruiz-Herguido C, López ME, Inglés-Esteve J, Riera L, Tipping A, Enver T, Dzierzak E, Gridley T, Espinosa L, Bigas A. Impaired embryonic haematopoiesis yet normal arterial development in the absence of the Notch ligand Jagged1. EMBO J 2008; 27:1886-95. [PMID: 18528438 PMCID: PMC2486417 DOI: 10.1038/emboj.2008.113] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2008] [Accepted: 05/14/2008] [Indexed: 11/08/2022] Open
Abstract
Specific deletion of Notch1 and RBPjkappa in the mouse results in abrogation of definitive haematopoiesis concomitant with the loss of arterial identity at embryonic stage. As prior arterial determination is likely to be required for the generation of embryonic haematopoiesis, it is difficult to establish the specific haematopoietic role of Notch in these mutants. By analysing different Notch-ligand-null embryos, we now show that Jagged1 is not required for the establishment of the arterial fate but it is required for the correct execution of the definitive haematopoietic programme, including expression of GATA2 in the dorsal aorta. Moreover, successful haematopoietic rescue of the Jagged1-null AGM cells was obtained by culturing them with Jagged1-expressing stromal cells or by lentiviral-mediated transduction of the GATA2 gene. Taken together, our results indicate that Jagged1-mediated activation of Notch1 is responsible for regulating GATA2 expression in the AGM, which in turn is essential for definitive haematopoiesis in the mouse.
Collapse
Affiliation(s)
- Àlex Robert-Moreno
- Centre Oncologia Molecular, IDIBELL, Gran Via km 2.7 Hospitalet, Barcelona, Spain
- These authors contributed equally to this work
| | - Jordi Guiu
- Centre Oncologia Molecular, IDIBELL, Gran Via km 2.7 Hospitalet, Barcelona, Spain
- These authors contributed equally to this work
| | | | - M Eugenia López
- Centre Oncologia Molecular, IDIBELL, Gran Via km 2.7 Hospitalet, Barcelona, Spain
| | - Julia Inglés-Esteve
- Centre Oncologia Molecular, IDIBELL, Gran Via km 2.7 Hospitalet, Barcelona, Spain
| | - Lluis Riera
- Centre Oncologia Molecular, IDIBELL, Gran Via km 2.7 Hospitalet, Barcelona, Spain
| | - Alex Tipping
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Tariq Enver
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Elaine Dzierzak
- Department of Cell Biology and Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Lluis Espinosa
- Centre Oncologia Molecular, IDIBELL, Gran Via km 2.7 Hospitalet, Barcelona, Spain
- These authors contributed equally to this work
| | - Anna Bigas
- Centre Oncologia Molecular, IDIBELL, Gran Via km 2.7 Hospitalet, Barcelona, Spain
- These authors contributed equally to this work
| |
Collapse
|