1
|
Khan AA, James D, Andresen V, Atkey J, Bradbury R, Cargo C, Dillon R, Gjertsen BT, Goldstone AR, Leach R, Lock D, Narayanan M, Russell N, Verigou EA, Green S, Fielding AK, Falini B. Pre-B acute lymphoblastic leukemia presenting with NPM1 and FLT3 mutations. Am J Hematol 2024; 99:2018-2022. [PMID: 38567798 DOI: 10.1002/ajh.27301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/23/2024] [Accepted: 03/11/2024] [Indexed: 10/25/2024]
Affiliation(s)
- Alesia A Khan
- Hematological Malignancy Diagnostic Service, Leeds Teaching Hospitals, Leeds, UK
| | - Daniel James
- Centre for Blood Research, Hull University Teaching Hospitals NHS Trust, Hull, UK
| | - Vibeke Andresen
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Julie Atkey
- Hematological Malignancy Diagnostic Service, Leeds Teaching Hospitals, Leeds, UK
| | - Rachel Bradbury
- Hematological Malignancy Diagnostic Service, Leeds Teaching Hospitals, Leeds, UK
| | - Catherine Cargo
- Hematological Malignancy Diagnostic Service, Leeds Teaching Hospitals, Leeds, UK
| | - Richard Dillon
- Department of Medical and Molecular Genetics, Guy's and King's Hospitals, London, UK
| | - Bjørn Tore Gjertsen
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
| | - Antony R Goldstone
- Centre for Blood Research, Hull University Teaching Hospitals NHS Trust, Hull, UK
| | - Richard Leach
- Hematological Malignancy Diagnostic Service, Leeds Teaching Hospitals, Leeds, UK
| | - Daniel Lock
- Hematological Malignancy Diagnostic Service, Leeds Teaching Hospitals, Leeds, UK
| | - Mayanka Narayanan
- Centre for Blood Research, Hull University Teaching Hospitals NHS Trust, Hull, UK
| | - Nigel Russell
- Department of Medical and Molecular Genetics, Guy's and King's Hospitals, London, UK
| | - Eleni-Anna Verigou
- Centre for Blood Research, Hull University Teaching Hospitals NHS Trust, Hull, UK
| | - Simone Green
- Centre for Blood Research, Hull University Teaching Hospitals NHS Trust, Hull, UK
| | - Adele K Fielding
- Centre for Blood Research, Hull University Teaching Hospitals NHS Trust, Hull, UK
- Centre for Blood research, University of York, York, UK
| | - Brunangelo Falini
- Institute of Hematology and Center for Haemato-Oncological research (CREO), University of Perugia and Santa Maria della Misericordia Hospital, Perugia, Italy
| |
Collapse
|
2
|
NPM 1 Mutations in AML-The Landscape in 2023. Cancers (Basel) 2023; 15:cancers15041177. [PMID: 36831522 PMCID: PMC9954410 DOI: 10.3390/cancers15041177] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/04/2023] [Accepted: 02/07/2023] [Indexed: 02/15/2023] Open
Abstract
Acute myeloid leukemia (AML) represents 80% of acute leukemia in adults and is characterized by clonal expansion of hematopoietic stem cells secondary to genomic mutations, rendering a selective growth advantage to the mutant clones. NPM1mut is found in around 30% of AML and clinically presents with leukocytosis, high blast percentage and extramedullary involvement. Considered as a "gate-keeper" mutation, NPM1mut appears to be a "first hit" in the process of leukemogenesis and development of overt leukemia. Commonly associated with other mutations (e.g., FLT 3, DNMT3A, TET2, SF3B1), NPM1 mutation in AML has an important role in diagnosis, prognosis, treatment and post-treatment monitoring. Several novel therapies targeting NPM1 are being developed in various clinical phases with demonstration of efficacy. In this review, we summarize the pathophysiology of the NPM1 gene mutation in AML, clinical implications and the novel targeted therapies to date.
Collapse
|
3
|
Sakthivel D, Brown-Suedel A, Bouchier-Hayes L. The role of the nucleolus in regulating the cell cycle and the DNA damage response. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 135:203-241. [PMID: 37061332 DOI: 10.1016/bs.apcsb.2023.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
The nucleolus has long been perceived as the site for ribosome biogenesis, but numerous studies suggest that the nucleolus carefully sequesters crucial proteins involved in multiple cellular functions. Among these, the role of nucleolus in cell cycle regulation is the most evident. The nucleolus is the first responder of growth-related signals to mediate normal cell cycle progression. The nucleolus also senses different cellular stress insults by activating diverse pathways that arrest the cell cycle, promote DNA repair, or initiate apoptosis. Here, we review the emerging concepts on how the ribosomal and nonribosomal nucleolar proteins mediate such cellular effects.
Collapse
|
4
|
Nucleophosmin 1 Mutations in Acute Myeloid Leukemia. Genes (Basel) 2020; 11:genes11060649. [PMID: 32545659 PMCID: PMC7348733 DOI: 10.3390/genes11060649] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/06/2020] [Accepted: 06/09/2020] [Indexed: 12/16/2022] Open
Abstract
Nucleophosmin (NPM1) is a ubiquitously expressed nucleolar protein involved in ribosome biogenesis, the maintenance of genomic integrity and the regulation of the ARF-p53 tumor-suppressor pathway among multiple other functions. Mutations in the corresponding gene cause a cytoplasmic dislocation of the NPM1 protein. These mutations are unique to acute myeloid leukemia (AML), a disease characterized by clonal expansion, impaired differentiation and the proliferation of myeloid cells in the bone marrow. Despite our improved understanding of NPM1 mutations and their consequences, the underlying leukemia pathogenesis is still unclear. Recent studies that focused on dysregulated gene expression in AML with mutated NPM1 have shed more light into these mechanisms. In this article, we review the current evidence on normal functions of NPM1 and aberrant functioning in AML, and highlight investigational strategies targeting these mutations.
Collapse
|
5
|
Nagy Á, Ősz Á, Budczies J, Krizsán S, Szombath G, Demeter J, Bödör C, Győrffy B. Elevated HOX gene expression in acute myeloid leukemia is associated with NPM1 mutations and poor survival. J Adv Res 2019; 20:105-116. [PMID: 31333881 PMCID: PMC6614546 DOI: 10.1016/j.jare.2019.05.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/27/2019] [Accepted: 05/28/2019] [Indexed: 12/20/2022] Open
Abstract
Acute myeloid leukemia (AML) is a clonal disorder of hematopoietic progenitor cells and the most common malignant myeloid disorder in adults. Several gene mutations such as in NPM1 (nucleophosmin 1) are involved in the pathogenesis and progression of AML. The aim of this study was to identify genes whose expression is associated with driver mutations and survival outcome. Genotype data (somatic mutations) and gene expression data including RNA-seq, microarray, and qPCR data were used for the analysis. Multiple datasets were utilized as training sets (GSE6891, TCGA, and GSE1159). A new clinical sample cohort (Semmelweis set) was established for in vitro validation. Wilcoxon analysis was used to identify genes with expression alterations between the mutant and wild type samples. Cox regression analysis was performed to examine the association between gene expression and survival outcome. Data analysis was performed in the R statistical environment. Eighty-five genes were identified with significantly altered expression when comparing NPM1 mutant and wild type patient groups in the GSE6891 set. Additional training sets were used as a filter to condense the six most significant genes associated with NPM1 mutations. Then, the expression changes of these six genes were confirmed in the Semmelweis set: HOXA5 (P = 3.06E-12, FC = 8.3), HOXA10 (P = 2.44E-09, FC = 3.3), HOXB5 (P = 1.86E-13, FC = 37), MEIS1 (P = 9.82E-10, FC = 4.4), PBX3 (P = 1.03E-13, FC = 5.4) and ITM2A (P = 0.004, FC = 0.4). Cox regression analysis showed that higher expression of these genes - with the exception of ITM2A - was associated with worse overall survival. Higher expression of the HOX genes was identified in tumors harboring NPM1 gene mutations by computationally linking genotype and gene expression. In vitro validation of these genes supports their potential therapeutic application in AML.
Collapse
Key Words
- AML, acute myeloid leukemia
- Acute myeloid leukemia
- Clinical samples
- FAB classification, French–American–British classification
- FC, fold change
- Gene expression
- HOX genes
- HOX, homeobox
- HR, hazard ratio
- ITD, internal tandem duplication
- MEIS, myeloid ecotropic viral integration site
- Mutation
- NCBI GEO, National Center for Biotechnology Gene expression Omnibus
- OS, overall survival
- PBX, pre-B-cell leukemia homeobox
- Survival
- TCGA, The Cancer Genome Atlas
- WHO, World Health Organization
- qPCR, quantitative polymerase chain reaction
Collapse
Affiliation(s)
- Ádám Nagy
- MTA TTK Lendület Cancer Biomarker Research Group, Hungarian Academy of Sciences Research Centre for Natural Sciences, Institute of Enzymology, Magyar Tudósok körútja 2, 1117 Budapest, Hungary.,Semmelweis University 2nd Dept. of Pediatrics, Tűzoltó utca 7-9, 1094 Budapest, Hungary
| | - Ágnes Ősz
- MTA TTK Lendület Cancer Biomarker Research Group, Hungarian Academy of Sciences Research Centre for Natural Sciences, Institute of Enzymology, Magyar Tudósok körútja 2, 1117 Budapest, Hungary.,Semmelweis University 2nd Dept. of Pediatrics, Tűzoltó utca 7-9, 1094 Budapest, Hungary
| | - Jan Budczies
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Szilvia Krizsán
- MTA-SE Lendület Molecular Oncohematology Research Group, 1st Department of Pathology, and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Gergely Szombath
- 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Judit Demeter
- 1st Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Csaba Bödör
- MTA-SE Lendület Molecular Oncohematology Research Group, 1st Department of Pathology, and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, Hungarian Academy of Sciences Research Centre for Natural Sciences, Institute of Enzymology, Magyar Tudósok körútja 2, 1117 Budapest, Hungary.,Semmelweis University 2nd Dept. of Pediatrics, Tűzoltó utca 7-9, 1094 Budapest, Hungary
| |
Collapse
|
6
|
Gu X, Ebrahem Q, Mahfouz RZ, Hasipek M, Enane F, Radivoyevitch T, Rapin N, Przychodzen B, Hu Z, Balusu R, Cotta CV, Wald D, Argueta C, Landesman Y, Martelli MP, Falini B, Carraway H, Porse BT, Maciejewski J, Jha BK, Saunthararajah Y. Leukemogenic nucleophosmin mutation disrupts the transcription factor hub that regulates granulomonocytic fates. J Clin Invest 2018; 128:4260-4279. [PMID: 30015632 PMCID: PMC6159976 DOI: 10.1172/jci97117] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 07/10/2018] [Indexed: 12/23/2022] Open
Abstract
Nucleophosmin (NPM1) is among the most frequently mutated genes in acute myeloid leukemia (AML). It is not known, however, how the resulting oncoprotein mutant NPM1 is leukemogenic. To reveal the cellular machinery in which NPM1 participates in myeloid cells, we analyzed the endogenous NPM1 protein interactome by mass spectrometry and discovered abundant amounts of the master transcription factor driver of monocyte lineage differentiation PU.1 (also known as SPI1). Mutant NPM1, which aberrantly accumulates in cytoplasm, dislocated PU.1 into cytoplasm with it. CEBPA and RUNX1, the master transcription factors that collaborate with PU.1 to activate granulomonocytic lineage fates, remained nuclear; but without PU.1, their coregulator interactions were toggled from coactivators to corepressors, repressing instead of activating more than 500 granulocyte and monocyte terminal differentiation genes. An inhibitor of nuclear export, selinexor, by locking mutant NPM1/PU.1 in the nucleus, activated terminal monocytic fates. Direct depletion of the corepressor DNA methyltransferase 1 (DNMT1) from the CEBPA/RUNX1 protein interactome using the clinical drug decitabine activated terminal granulocytic fates. Together, these noncytotoxic treatments extended survival by more than 160 days versus vehicle in a patient-derived xenotransplant model of NPM1/FLT3-mutated AML. In sum, mutant NPM1 represses monocyte and granulocyte terminal differentiation by disrupting PU.1/CEBPA/RUNX1 collaboration, a transforming action that can be reversed by pharmacodynamically directed dosing of clinical small molecules.
Collapse
Affiliation(s)
- Xiaorong Gu
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Quteba Ebrahem
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Reda Z. Mahfouz
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Metis Hasipek
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Francis Enane
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Tomas Radivoyevitch
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Nicolas Rapin
- The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Stem Cell Biology, DanStem, and Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bartlomiej Przychodzen
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Zhenbo Hu
- Department of Hematology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Ramesh Balusu
- Department of Internal Medicine, Division of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Claudiu V. Cotta
- Department of Clinical Pathology, Tomsich Pathology Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - David Wald
- Department of Clinical Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | | | | | - Maria Paola Martelli
- Institute of Hematology, Center for Research in Hematology-Oncology (CREO), University of Perugia, Perugia, Italy
| | - Brunangelo Falini
- Institute of Hematology, Center for Research in Hematology-Oncology (CREO), University of Perugia, Perugia, Italy
| | - Hetty Carraway
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Bo T. Porse
- The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Stem Cell Biology, DanStem, and Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jaroslaw Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Babal K. Jha
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Yogen Saunthararajah
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
7
|
Heath EM, Chan SM, Minden MD, Murphy T, Shlush LI, Schimmer AD. Biological and clinical consequences of NPM1 mutations in AML. Leukemia 2017; 31:798-807. [PMID: 28111462 DOI: 10.1038/leu.2017.30] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 01/09/2017] [Accepted: 01/13/2017] [Indexed: 12/16/2022]
Abstract
Acute myeloid leukemia (AML) is characterized by accumulation of myeloid cells in the bone marrow because of impaired differentiation and proliferation, resulting in hematopoietic insufficiency. NPM1 is one of the most commonly mutated genes in AML, present in 20-30% of cases. Mutations in NPM1 represent a distinct entity in the World Health Organization (WHO) classification and commonly indicate a better risk prognosis. In this review, we discuss the many functions of NPM1, the consequence of mutations in NPM1 and possible mechanisms through which mutations lead to leukemogenesis. We also discuss clinical consequences of mutations, associated gene expression patterns and the role of NPM1 mutations in informing prognosis and therapeutic decisions and predicting relapse in AML.
Collapse
Affiliation(s)
- E M Heath
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Ontario, Canada
| | - S M Chan
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - M D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Ontario, Canada
| | - T Murphy
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Ontario, Canada
| | - L I Shlush
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - A D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Ontario, Canada
| |
Collapse
|
8
|
von Bonin M, Wermke M, Cosgun KN, Thiede C, Bornhauser M, Wagemaker G, Waskow C. In vivo expansion of co-transplanted T cells impacts on tumor re-initiating activity of human acute myeloid leukemia in NSG mice. PLoS One 2013; 8:e60680. [PMID: 23585844 PMCID: PMC3621959 DOI: 10.1371/journal.pone.0060680] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 03/01/2013] [Indexed: 01/19/2023] Open
Abstract
Human cells from acute myeloid leukemia (AML) patients are frequently transplanted into immune-compromised mouse strains to provide an in vivo environment for studies on the biology of the disease. Since frequencies of leukemia re-initiating cells are low and a unique cell surface phenotype that includes all tumor re-initiating activity remains unknown, the underlying mechanisms leading to limitations in the xenotransplantation assay need to be understood and overcome to obtain robust engraftment of AML-containing samples. We report here that in the NSG xenotransplantation assay, the large majority of mononucleated cells from patients with AML fail to establish a reproducible myeloid engraftment despite high donor chimerism. Instead, donor-derived cells mainly consist of polyclonal disease-unrelated expanded co-transplanted human T lymphocytes that induce xenogeneic graft versus host disease and mask the engraftment of human AML in mice. Engraftment of mainly myeloid cell types can be enforced by the prevention of T cell expansion through the depletion of lymphocytes from the graft prior transplantation.
Collapse
Affiliation(s)
- Malte von Bonin
- Regeneration in Hematopoiesis, Center for Regenerative Therapies Dresden – CRTD, DFG Research Center and Cluster of Excellence, Technische Universität Dresden, Dresden, Germany
- Medizinische Klinik und Poliklinik 1, Universitätsklinikum Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Martin Wermke
- Medizinische Klinik und Poliklinik 1, Universitätsklinikum Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Kadriye Nehir Cosgun
- Regeneration in Hematopoiesis, Center for Regenerative Therapies Dresden – CRTD, DFG Research Center and Cluster of Excellence, Technische Universität Dresden, Dresden, Germany
| | - Christian Thiede
- Medizinische Klinik und Poliklinik 1, Universitätsklinikum Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Martin Bornhauser
- Medizinische Klinik und Poliklinik 1, Universitätsklinikum Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Gerard Wagemaker
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Claudia Waskow
- Regeneration in Hematopoiesis, Center for Regenerative Therapies Dresden – CRTD, DFG Research Center and Cluster of Excellence, Technische Universität Dresden, Dresden, Germany
- * E-mail:
| |
Collapse
|
9
|
Dvorakova D, Racil Z, Borsky M, Robesova B, Jeziskova I, Razga F, Lengerova M, Mayer J. Clonal heterogeneity in patients with cytogenetically normal acute myeloid leukemia withNPM1mutations. Leuk Lymphoma 2012; 54:1056-60. [DOI: 10.3109/10428194.2012.734618] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
10
|
The Multifunctional Nucleolar Protein Nucleophosmin/NPM/B23 and the Nucleoplasmin Family of Proteins. THE NUCLEOLUS 2011. [PMCID: PMC7121557 DOI: 10.1007/978-1-4614-0514-6_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The nucleophosmin (NPM)/nucleoplasmin family of nuclear chaperones has three members: NPM1, NPM2, and NPM3. Nuclear chaperones serve to ensure proper assembly of nucleosomes and proper formation of higher order structures of chromatin. In fact, this family of proteins has such diverse functions in cellular processes such as chromatin remodeling, ribosome biogenesis, genome stability, centrosome replication, cell cycle, transcriptional regulation, apoptosis, and tumor suppression. Of the members of this family, NPM1 is the most studied and is the main focus of this review. NPM2 and NPM3 are less well characterized, and are also discussed wherever appropriate. The structure–function relationship of NPM proteins has largely been worked out. Other than the many processes in which NPM1 takes part, the major interest comes from its involvement in human cancers, particularly acute myeloid leukemia (AML). Its significance stems from the fact that AML with mutated NPM1 accounts for ∼30% of all AML cases and usually has good prognosis. Its clinical importance also comes from its involvement in virus replication, particularly in the era of outbreaks of infectious diseases.
Collapse
|
11
|
CD34+ cells from AML with mutated NPM1 harbor cytoplasmic mutated nucleophosmin and generate leukemia in immunocompromised mice. Blood 2010; 116:3907-22. [DOI: 10.1182/blood-2009-08-238899] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Abstract
Acute myeloid leukemia (AML) with mutated NPM1 shows distinctive biologic and clinical features, including absent/low CD34 expression, the significance of which remains unclear. Therefore, we analyzed CD34+ cells from 41 NPM1-mutated AML. At flow cytometry, 31 of 41 samples contained less than 10% cells showing low intensity CD34 positivity and variable expression of CD38. Mutational analysis and/or Western blotting of purified CD34+ cells from 17 patients revealed NPM1-mutated gene and/or protein in all. Immunohistochemistry of trephine bone marrow biopsies and/or flow cytometry proved CD34+ leukemia cells from NPM1-mutated AML had aberrant nucleophosmin expression in cytoplasm. NPM1-mutated gene and/or protein was also confirmed in a CD34+ subfraction exhibiting the phenotype (CD34+/CD38−/CD123+/CD33+/CD90−) of leukemic stem cells. When transplanted into immunocompromised mice, CD34+ cells generated a leukemia recapitulating, both morphologically and immunohistochemically (aberrant cytoplasmic nucleophosmin, CD34 negativity), the original patient's disease. These results indicate that the CD34+ fraction in NPM1-mutated AML belongs to the leukemic clone and contains NPM1-mutated cells exhibiting properties typical of leukemia-initiating cells. CD34− cells from few cases (2/15) also showed significant leukemia-initiating cell potential in immunocompromised mice. This study provides further evidence that NPM1 mutation is a founder genetic lesion and has potential implications for the cell-of-origin and targeted therapy of NPM1-mutated AML.
Collapse
|
12
|
Oelschlaegel U, Koch S, Mohr B, Schaich M, Falini B, Ehninger G, Thiede C. Rapid flow cytometric detection of aberrant cytoplasmic localization of nucleophosmin (NPMc) indicating mutant NPM1 gene in acute myeloid leukemia. Leukemia 2010; 24:1813-6. [DOI: 10.1038/leu.2010.178] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
13
|
Multilineage dysplasia has no impact on biologic, clinicopathologic, and prognostic features of AML with mutated nucleophosmin (NPM1). Blood 2010; 115:3776-86. [DOI: 10.1182/blood-2009-08-240457] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
NPM1-mutated acute myeloid leukemia (AML) is a provisional entity in the 2008 World Health Organization (WHO) classification of myeloid neoplasms. The significance of multilineage dysplasia (MLD) in NPM1-mutated AML is unclear. Thus, in the 2008 WHO classification, NPM1-mutated AML with MLD is classified as AML with myelodysplasia (MD)–related changes (MRCs). We evaluated morphologically 318 NPM1-mutated AML patients and found MLD in 23.3%. Except for a male predominance and a lower fms-related tyrosine kinase 3–internal tandem duplication (FLT3-ITD) incidence in the MLD+ group, no differences were observed in age, sex, cytogenetics, and FLT3-–tyrosine kinase domain between NPM1-mutated AML with and without MLD. NPM1-mutated AML with and without MLD showed overlapping immunophenotype (CD34 negativity) and gene expression profile (CD34 down-regulation, HOX genes up-regulation). Moreover, overall and event-free survival did not differ among NPM1-mutated AML patients independently of whether they were MLD+ or MLD−, the NPM1-mutated/FLT3-ITD negative genotype showing the better prognosis. Lack of MLD impact on survival was confirmed by multivariate analysis that highlighted FLT3-ITD as the only significant prognostic parameter in NPM1-mutated AML. Our findings indicate that NPM1 mutations rather than MLD dictate the distinctive features of NPM1-mutated AML. Thus, irrespective of MLD, NPM1-mutated AML represents one disease entity clearly distinct from AML with MRCs.
Collapse
|
14
|
Leukemia-initiating cells from some acute myeloid leukemia patients with mutated nucleophosmin reside in the CD34(-) fraction. Blood 2010; 115:1976-84. [PMID: 20053758 DOI: 10.1182/blood-2009-02-206565] [Citation(s) in RCA: 276] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Leukemia-initiating cells (LICs) in acute myeloid leukemia (AML) are believed to be restricted to the CD34(+) fraction. However, one of the most frequently mutated genes in AML is nucleophosmin (NPM), and this is associated with low CD34 expression. We, therefore, investigated whether NPM-mutated AMLs have LICs restricted to the CD34(+) fraction. We transplanted sorted fractions of primary NPM-mutated AML into immunodeficient mice to establish which fractions initiate leukemia. Approximately one-half of cases had LICs exclusively within the CD34(-) fraction, whereas the CD34(+) fraction contained normal multilineage hematopoietic repopulating cells. Most of the remaining cases had LICs in both CD34(+) and CD34(-) fractions. When samples were sorted based on CD34 and CD38 expression, multiple fractions initiated leukemia in primary and secondary recipients. The data indicate that the phenotype of LICs is more heterogeneous than previously realized and can vary even within a single sample. This feature of LICs may make them particularly difficult to eradicate using therapies targeted against surface antigens.
Collapse
|
15
|
Rau R, Brown P. Nucleophosmin (NPM1) mutations in adult and childhood acute myeloid leukaemia: towards definition of a new leukaemia entity. Hematol Oncol 2009; 27:171-81. [PMID: 19569254 DOI: 10.1002/hon.904] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Nucleophosmin (NPM) is a ubiquitously expressed chaperone protein that shuttles rapidly between the nucleus and cytoplasm, but predominantly resides in the nucleolus. It plays key roles in ribosome biogenesis, centrosome duplication, genomic stability, cell cycle progression and apoptosis. Somatic mutations in exon 12 of the NPM gene (NPM1) are the most frequent genetic abnormality in adult acute myeloid leukaemia (AML), found in approximately 35% of all cases and up to 60% of patients with normal karyotype (NK) AML. In children, NPM1 mutations are far less frequent, occurring in 8-10% of all AML cases, and in approximately 25% of those with a NK. NPM1 mutations lead to aberrant localization of the NPM protein into the cytoplasm, thus the designation, NPMc+ AML. NPMc+ AML is seen predominantly in patients with a NK and is essentially mutually exclusive of recurrent chromosomal translocations. Patients with NPM1 mutations are twice as likely as those who lack an NPM1 mutation to also have a FMS-like tyrosine kinase (FLT3) internal tandem duplication (ITD) mutation. NPMc+ AML is also characterized by a unique gene expression signature and microRNA signature. NPMc+ AML has important prognostic significance, as NPMc+ AML, in the absence of a coexisting FLT3-ITD mutation, is associated with a favourable outcome. NPM1 mutations have also shown great stability during disease evolution, and therefore represent a possible marker for minimal residual disease detection. Given its distinctive biologic and clinical features and its clear clinical relevance, NPMc+ AML is included as a provisional entity in the 2008 WHO classifications. There is still much to be learned about this genetic alteration, including its exact role in leukaemogenesis, how it interacts with other mutations and why it confers a more favourable prognosis. Further, it represents a potential therapeutic target warranting research aimed at identifying novel small molecules with activity in NPMc+ AML.
Collapse
Affiliation(s)
- Rachel Rau
- Departments of Oncology and Pediatrics, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | | |
Collapse
|
16
|
Falini B, Sportoletti P, Martelli MP. Acute myeloid leukemia with mutated NPM1: diagnosis, prognosis and therapeutic perspectives. Curr Opin Oncol 2009; 21:573-81. [DOI: 10.1097/cco.0b013e3283313dfa] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
17
|
Facchetti F, Pileri SA, Agostinelli C, Martelli MP, Paulli M, Venditti A, Martelli MF, Falini B. Cytoplasmic nucleophosmin is not detected in blastic plasmacytoid dendritic cell neoplasm. Haematologica 2008; 94:285-8. [PMID: 19066330 DOI: 10.3324/haematol.13855] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Acute myeloid leukemia carrying cytoplasmic mutated nucleophosmin (NPMc(+) AML) and blastic plasmacytoid dendritic cell neoplasm have been included as new entities in the 4(th) edition (2008) WHO classification of myeloid neoplasms. These conditions may show clinical and pathological overlapping features (leukemic and skin involvement, and expression of macrophage markers). In this study, we provide evidence that aberrant cytoplasmic dislocation of nucleophosmin - the immunohistochemical surrogate for NPM1 mutations - allows the two entities to be genetically separated. In fact, nucleophosmin is consistently cytoplasmic in NPMc(+) AML (because of the presence of NPM1 mutations), whilst it is nucleus-restricted (predictive of a germline NPM1 gene) in blastic plasmacytoid dendritic cell neoplasm. Our results clearly point cytoplasmic nucleophosmin (a full predictor of NPM1 mutations) as a new marker for distinguishing NPMc(+) AML and blastic plasmacytoid dendritic cell neoplasm, further clarify the cell of origin of NPMc(+) AML, and justify the inclusion of these pathological conditions as separate entities in the new WHO classification.
Collapse
Affiliation(s)
- Fabio Facchetti
- Institute of Pathology, Spedali Civili, University of Brescia, Brescia
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Garzon R, Garofalo M, Martelli MP, Briesewitz R, Wang L, Fernandez-Cymering C, Volinia S, Liu CG, Schnittger S, Haferlach T, Liso A, Diverio D, Mancini M, Meloni G, Foa R, Martelli MF, Mecucci C, Croce CM, Falini B. Distinctive microRNA signature of acute myeloid leukemia bearing cytoplasmic mutated nucleophosmin. Proc Natl Acad Sci U S A 2008; 105:3945-3950. [PMID: 18308931 PMCID: PMC2268779 DOI: 10.1073/pnas.0800135105] [Citation(s) in RCA: 384] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2007] [Indexed: 12/16/2022] Open
Abstract
Acute myeloid leukemia (AML) carrying NPM1 mutations and cytoplasmic nucleophosmin (NPMc+ AML) accounts for about one-third of adult AML and shows distinct features, including a unique gene expression profile. MicroRNAs (miRNAs) are small noncoding RNAs of 19-25 nucleotides in length that have been linked to the development of cancer. Here, we investigated the role of miRNAs in the biology of NPMc+ AML. The miRNA expression was evaluated in 85 adult de novo AML patients characterized for subcellular localization/mutation status of NPM1 and FLT3 mutations using a custom microarray platform. Data were analyzed by using univariate t test within BRB tools. We identified a strong miRNA signature that distinguishes NPMc+ mutated (n = 55) from the cytoplasmic-negative (NPM1 unmutated) cases (n = 30) and includes the up-regulation of miR-10a, miR-10b, several let-7 and miR-29 family members. Many of the down-regulated miRNAs including miR-204 and miR-128a are predicted to target several HOX genes. Indeed, we confirmed that miR-204 targets HOXA10 and MEIS1, suggesting that the HOX up-regulation observed in NPMc+ AML may be due in part by loss of HOX regulators-miRNAs. FLT3-ITD+ samples were characterized by up-regulation of miR-155. Further experiments demonstrated that the up-regulation of miR-155 was independent from FLT3 signaling. Our results identify a unique miRNA signature associated with NPMc+ AML and provide evidence that support a role for miRNAs in the regulation of HOX genes in this leukemia subtype. Moreover, we found that miR-155 was strongly but independently associated with FLT3-ITD mutations.
Collapse
Affiliation(s)
| | - Michela Garofalo
- Molecular Virology and Human Genetics, Comprehensive Cancer Center, and
| | | | - Roger Briesewitz
- College of Pharmacology, Ohio State University, Columbus, OH 43221
| | - Lisheng Wang
- College of Pharmacology, Ohio State University, Columbus, OH 43221
| | | | - Stefano Volinia
- Molecular Virology and Human Genetics, Comprehensive Cancer Center, and
| | - Chang-Gong Liu
- Molecular Virology and Human Genetics, Comprehensive Cancer Center, and
| | | | | | - Arcangelo Liso
- Institute of Hematology, University of Foggia, 71020 Foggia, Italy; and
| | - Daniela Diverio
- **Institute of Hematology, University “La Sapienza,” 0185 Rome, Italy
| | - Marco Mancini
- **Institute of Hematology, University “La Sapienza,” 0185 Rome, Italy
| | - Giovanna Meloni
- **Institute of Hematology, University “La Sapienza,” 0185 Rome, Italy
| | - Robin Foa
- **Institute of Hematology, University “La Sapienza,” 0185 Rome, Italy
| | | | - Cristina Mecucci
- Institute of Hematology, University of Perugia, 6100 Perugia, Italy
| | - Carlo M. Croce
- Molecular Virology and Human Genetics, Comprehensive Cancer Center, and
| | | |
Collapse
|
19
|
Liso A, Bogliolo A, Freschi V, Martelli MP, Pileri SA, Santodirocco M, Bolli N, Martelli MF, Falini B. In human genome, generation of a nuclear export signal through duplication appears unique to nucleophosmin (NPM1) mutations and is restricted to AML. Leukemia 2007; 22:1285-9. [DOI: 10.1038/sj.leu.2405045] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|