1
|
Ulloa-Aguilar JM, Herrera Moro Huitron L, Benítez-Zeferino RY, Cerna-Cortes JF, García-Cordero J, León-Reyes G, Guzman-Bautista ER, Farfan-Morales CN, Reyes-Ruiz JM, Miranda-Labra RU, De Jesús-González LA, León-Juárez M. The Nucleolus and Its Interactions with Viral Proteins Required for Successful Infection. Cells 2024; 13:1591. [PMID: 39329772 PMCID: PMC11430610 DOI: 10.3390/cells13181591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
Nuclear bodies are structures in eukaryotic cells that lack a plasma membrane and are considered protein condensates, DNA, or RNA molecules. Known nuclear bodies include the nucleolus, Cajal bodies, and promyelocytic leukemia nuclear bodies. These bodies are involved in the concentration, exclusion, sequestration, assembly, modification, and recycling of specific components involved in the regulation of ribosome biogenesis, RNA transcription, and RNA processing. Additionally, nuclear bodies have been shown to participate in cellular processes such as the regulation of transcription of the cell cycle, mitosis, apoptosis, and the cellular stress response. The dynamics and functions of these bodies depend on the state of the cell. It is now known that both DNA and RNA viruses can direct their proteins to nuclear bodies, causing alterations in their composition, dynamics, and functions. Although many of these mechanisms are still under investigation, it is well known that the interaction between viral and nuclear body proteins is necessary for the success of the viral infection cycle. In this review, we concisely describe the interaction between viral and nuclear body proteins. Furthermore, we focus on the role of the nucleolus in RNA virus infections. Finally, we discuss the possible implications of the interaction of viral proteins on cellular transcription and the formation/degradation of non-coding RNAs.
Collapse
Affiliation(s)
- José Manuel Ulloa-Aguilar
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (J.M.U.-A.); (L.H.M.H.); (R.Y.B.-Z.); (E.R.G.-B.)
- Posgrado en Biología Experimental, Departamento de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09310, Mexico
| | - Luis Herrera Moro Huitron
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (J.M.U.-A.); (L.H.M.H.); (R.Y.B.-Z.); (E.R.G.-B.)
- Laboratorio de Microbiología Molecular, Departamento de Microbiología, Escuela Nacional de Ciencias Biologícas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Rocío Yazmin Benítez-Zeferino
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (J.M.U.-A.); (L.H.M.H.); (R.Y.B.-Z.); (E.R.G.-B.)
- Laboratorio de Microbiología Molecular, Departamento de Microbiología, Escuela Nacional de Ciencias Biologícas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Jorge Francisco Cerna-Cortes
- Laboratorio de Microbiología Molecular, Departamento de Microbiología, Escuela Nacional de Ciencias Biologícas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Julio García-Cordero
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico;
| | - Guadalupe León-Reyes
- Laboratorio de Nutrigenética y Nutrigenómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico;
| | - Edgar Rodrigo Guzman-Bautista
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (J.M.U.-A.); (L.H.M.H.); (R.Y.B.-Z.); (E.R.G.-B.)
| | - Carlos Noe Farfan-Morales
- Departamento de Ciencias Naturales, Universidad Autonoma Metropolitana (UAM), Unidad Cuajimalpa, Mexico City 05348, Mexico;
| | - José Manuel Reyes-Ruiz
- Centro Médico Nacional “Adolfo Ruiz Cortines”, Instituto Mexicano del Seguro Social (IMSS), Veracruz 91897, Mexico;
| | - Roxana U. Miranda-Labra
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09310, Mexico;
| | | | - Moises León-Juárez
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (J.M.U.-A.); (L.H.M.H.); (R.Y.B.-Z.); (E.R.G.-B.)
| |
Collapse
|
2
|
Buggiani J, Meinnel T, Giglione C, Frottin F. Advances in nuclear proteostasis of metazoans. Biochimie 2024:S0300-9084(24)00081-6. [PMID: 38642824 DOI: 10.1016/j.biochi.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 04/22/2024]
Abstract
The proteostasis network and associated protein quality control (PQC) mechanisms ensure proteome functionality and are essential for cell survival. A distinctive feature of eukaryotic cells is their high degree of compartmentalization, requiring specific and adapted proteostasis networks for each compartment. The nucleus, essential for maintaining the integrity of genetic information and gene transcription, is one such compartment. While PQC mechanisms have been investigated for decades in the cytoplasm and the endoplasmic reticulum, our knowledge of nuclear PQC pathways is only emerging. Recent developments in the field have underscored the importance of spatially managing aberrant proteins within the nucleus. Upon proteotoxic stress, misfolded proteins and PQC effectors accumulate in various nuclear membrane-less organelles. Beyond bringing together effectors and substrates, the biophysical properties of these organelles allow novel PQC functions. In this review, we explore the specificity of the nuclear compartment, the effectors of the nuclear proteostasis network, and the PQC roles of nuclear membrane-less organelles in metazoans.
Collapse
Affiliation(s)
- Julia Buggiani
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CEA, CNRS, Gif-sur-Yvette, France
| | - Thierry Meinnel
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CEA, CNRS, Gif-sur-Yvette, France
| | - Carmela Giglione
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CEA, CNRS, Gif-sur-Yvette, France
| | - Frédéric Frottin
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CEA, CNRS, Gif-sur-Yvette, France.
| |
Collapse
|
3
|
Volik PI, Kopeina GS, Zhivotovsky B, Zamaraev AV. Total recall: the role of PIDDosome components in neurodegeneration. Trends Mol Med 2023; 29:996-1013. [PMID: 37716905 DOI: 10.1016/j.molmed.2023.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/18/2023]
Abstract
The PIDDosome is a multiprotein complex that includes p53-induced protein with a death domain 1 (PIDD1), receptor-interacting protein-associated ICH-1/CED-3 homologous protein with a death domain (RAIDD), and caspase-2, the activation of which is driven by PIDDosome assembly. In addition to the key role of the PIDDosome in the regulation of cell differentiation, tissue homeostasis, and organogenesis and regeneration, caspase-2, RAIDD and PIDD1 engagement in neuronal development was shown. Here, we focus on the involvement of PIDDosome components in neurodegenerative disorders, including retinal neuropathies, different types of brain damage, and Alzheimer's disease (AD), Huntington's disease (HD), and Lewy body disease. We also discuss pathogenic variants of PIDD1, RAIDD, and caspase-2 that are associated with intellectual, behavioral, and psychological abnormalities, together with prospective PIDDosome inhibition strategies and their potential clinical application.
Collapse
Affiliation(s)
- Pavel I Volik
- Facuty of Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; Engelhardt Institute of Molecular Biology, RAS, 119991 Moscow, Russia
| | - Gelina S Kopeina
- Facuty of Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; Engelhardt Institute of Molecular Biology, RAS, 119991 Moscow, Russia
| | - Boris Zhivotovsky
- Facuty of Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; Engelhardt Institute of Molecular Biology, RAS, 119991 Moscow, Russia; Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177 Stockholm, Sweden.
| | - Alexey V Zamaraev
- Facuty of Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; Engelhardt Institute of Molecular Biology, RAS, 119991 Moscow, Russia.
| |
Collapse
|
4
|
Buneeva O, Kopylov A, Gnedenko O, Medvedeva M, Veselovsky A, Ivanov A, Zgoda V, Medvedev A. Proteomic Profiling of Mouse Brain Pyruvate Kinase Binding Proteins: A Hint for Moonlighting Functions of PKM1? Int J Mol Sci 2023; 24:ijms24087634. [PMID: 37108803 PMCID: PMC10143413 DOI: 10.3390/ijms24087634] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
Affinity-based proteomic profiling is widely used for the identification of proteins involved in the formation of various interactomes. Since protein-protein interactions (PPIs) reflect the role of particular proteins in the cell, identification of interaction partners for a protein of interest can reveal its function. The latter is especially important for the characterization of multifunctional proteins, which can play different roles in the cell. Pyruvate kinase (PK), a classical glycolytic enzyme catalyzing the last step of glycolysis, exists in four isoforms: PKM1, PKM2, PKL, and PKR. The enzyme isoform expressed in actively dividing cells, PKM2, exhibits many moonlighting (noncanonical) functions. In contrast to PKM2, PKM1, predominantly expressed in adult differentiated tissues, lacks well-documented moonlighting functions. However, certain evidence exists that it can also perform some functions unrelated to glycolysis. In order to evaluate protein partners, bound to PKM1, in this study we have combined affinity-based separation of mouse brain proteins with mass spectrometry identification. The highly purified PKM1 and a 32-mer synthetic peptide (PK peptide), sharing high sequence homology with the interface contact region of all PK isoforms, were used as the affinity ligands. This proteomic profiling resulted in the identification of specific and common proteins bound to both affinity ligands. Quantitative affinity binding to the affinity ligands of selected identified proteins was validated using a surface plasmon resonance (SPR) biosensor. Bioinformatic analysis has shown that the identified proteins, bound to both full-length PKM1 and the PK peptide, form a protein network (interactome). Some of these interactions are relevant for the moonlighting functions of PKM1. The proteomic dataset is available via ProteomeXchange with the identifier PXD041321.
Collapse
Affiliation(s)
- Olga Buneeva
- Institute of Biomedical Chemistry, 10 Pogodinskaya Street, Moscow 119121, Russia
| | - Arthur Kopylov
- Institute of Biomedical Chemistry, 10 Pogodinskaya Street, Moscow 119121, Russia
| | - Oksana Gnedenko
- Institute of Biomedical Chemistry, 10 Pogodinskaya Street, Moscow 119121, Russia
| | - Marina Medvedeva
- Department of Biochemistry, School of Biology, Moscow State University, Moscow 119991, Russia
| | - Alexander Veselovsky
- Institute of Biomedical Chemistry, 10 Pogodinskaya Street, Moscow 119121, Russia
| | - Alexis Ivanov
- Institute of Biomedical Chemistry, 10 Pogodinskaya Street, Moscow 119121, Russia
| | - Victor Zgoda
- Institute of Biomedical Chemistry, 10 Pogodinskaya Street, Moscow 119121, Russia
| | - Alexei Medvedev
- Institute of Biomedical Chemistry, 10 Pogodinskaya Street, Moscow 119121, Russia
| |
Collapse
|
5
|
Dörner K, Ruggeri C, Zemp I, Kutay U. Ribosome biogenesis factors-from names to functions. EMBO J 2023; 42:e112699. [PMID: 36762427 PMCID: PMC10068337 DOI: 10.15252/embj.2022112699] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/13/2022] [Accepted: 01/19/2023] [Indexed: 02/11/2023] Open
Abstract
The assembly of ribosomal subunits is a highly orchestrated process that involves a huge cohort of accessory factors. Most eukaryotic ribosome biogenesis factors were first identified by genetic screens and proteomic approaches of pre-ribosomal particles in Saccharomyces cerevisiae. Later, research on human ribosome synthesis not only demonstrated that the requirement for many of these factors is conserved in evolution, but also revealed the involvement of additional players, reflecting a more complex assembly pathway in mammalian cells. Yet, it remained a challenge for the field to assign a function to many of the identified factors and to reveal their molecular mode of action. Over the past decade, structural, biochemical, and cellular studies have largely filled this gap in knowledge and led to a detailed understanding of the molecular role that many of the players have during the stepwise process of ribosome maturation. Such detailed knowledge of the function of ribosome biogenesis factors will be key to further understand and better treat diseases linked to disturbed ribosome assembly, including ribosomopathies, as well as different types of cancer.
Collapse
Affiliation(s)
- Kerstin Dörner
- Department of Biology, Institute of Biochemistry, ETH Zurich, Zurich, Switzerland.,Molecular Life Sciences Ph.D. Program, Zurich, Switzerland
| | - Chiara Ruggeri
- Department of Biology, Institute of Biochemistry, ETH Zurich, Zurich, Switzerland.,RNA Biology Ph.D. Program, Zurich, Switzerland
| | - Ivo Zemp
- Department of Biology, Institute of Biochemistry, ETH Zurich, Zurich, Switzerland
| | - Ulrike Kutay
- Department of Biology, Institute of Biochemistry, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
6
|
Saluri M, Leppert A, Gese GV, Sahin C, Lama D, Kaldmäe M, Chen G, Elofsson A, Allison TM, Arsenian-Henriksson M, Johansson J, Lane DP, Hällberg BM, Landreh M. A "grappling hook" interaction connects self-assembly and chaperone activity of Nucleophosmin 1. PNAS NEXUS 2023; 2:pgac303. [PMID: 36743470 PMCID: PMC9896144 DOI: 10.1093/pnasnexus/pgac303] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023]
Abstract
How the self-assembly of partially disordered proteins generates functional compartments in the cytoplasm and particularly in the nucleus is poorly understood. Nucleophosmin 1 (NPM1) is an abundant nucleolar protein that forms large oligomers and undergoes liquid-liquid phase separation by binding RNA or ribosomal proteins. It provides the scaffold for ribosome assembly but also prevents protein aggregation as part of the cellular stress response. Here, we use aggregation assays and native mass spectrometry (MS) to examine the relationship between the self-assembly and chaperone activity of NPM1. We find that oligomerization of full-length NPM1 modulates its ability to retard amyloid formation in vitro. Machine learning-based structure prediction and cryo-electron microscopy reveal fuzzy interactions between the acidic disordered region and the C-terminal nucleotide-binding domain, which cross-link NPM1 pentamers into partially disordered oligomers. The addition of basic peptides results in a tighter association within the oligomers, reducing their capacity to prevent amyloid formation. Together, our findings show that NPM1 uses a "grappling hook" mechanism to form a network-like structure that traps aggregation-prone proteins. Nucleolar proteins and RNAs simultaneously modulate the association strength and chaperone activity, suggesting a mechanism by which nucleolar composition regulates the chaperone activity of NPM1.
Collapse
Affiliation(s)
- Mihkel Saluri
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet – Biomedicum, Solnavägen 9, 171 65 Solna, Stockholm, Sweden
| | | | | | - Cagla Sahin
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet – Biomedicum, Solnavägen 9, 171 65 Solna, Stockholm, Sweden,Structural Biology and NMR laboratory and the Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen, Denmark
| | - Dilraj Lama
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet – Biomedicum, Solnavägen 9, 171 65 Solna, Stockholm, Sweden
| | - Margit Kaldmäe
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet – Biomedicum, Solnavägen 9, 171 65 Solna, Stockholm, Sweden
| | - Gefei Chen
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57 Huddinge,, Sweden
| | - Arne Elofsson
- Science for Life Laboratory and Department of Biochemistry and Biophysics, Stockholm University, 114 19 Stockholm, Sweden
| | - Timothy M Allison
- Biomolecular Interaction Centre, School of Physical and Chemical Sciences, University of Canterbury, Upper Riccarton, Christchurch 8041, New Zealand
| | - Marie Arsenian-Henriksson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet – Biomedicum, Solnavägen 9, 171 65 Solna, Stockholm, Sweden
| | - Jan Johansson
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57 Huddinge,, Sweden
| | - David P Lane
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet – Biomedicum, Solnavägen 9, 171 65 Solna, Stockholm, Sweden
| | | | | |
Collapse
|
7
|
Ningombam A, Verma D, Kumar R, Singh J, Ali MS, Pandey AK, Singh I, Bakhshi S, Sharma A, Pushpam D, Palanichamy JK, Tanwar P, Singh AR, Chopra A. Prognostic relevance of NPM1, CEBPA, and FLT3 mutations in cytogenetically normal adult AML patients. AMERICAN JOURNAL OF BLOOD RESEARCH 2023; 13:28-43. [PMID: 36937459 PMCID: PMC10017593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 01/06/2023] [Indexed: 03/21/2023]
Abstract
BACKGROUND Acute myeloid leukemia with normal cytogenetics (CN-AML) is the largest group of AML patients with very heterogenous patient outcomes. The revised World Health Organization classification of the hematolymphoid tumours, 2022, has incorporated AML with Nucleophosphmin1 (NPM1) and CCAAT/enhancer binding protein-alpha (CEBPA) mutations as distinct entities. Despite the existing evidence of the prognostic relevance of FMS-like tyrosine kinase-3 internal tandem duplication (FLT3-ITD) in AML, it has not been included in the revised classification. METHOD In this prospective study, we determined the prevalence of NPM1, CEBPA, and FLT3 gene mutations in 151 de novo CN-AML adult patients (age ≥18 years) in a tertiary care hospital in north India. Additionally, the prognostic relevance of these mutations was also evaluated. RESULTS NPM1, FLT3-ITD, and CEBPA mutations were found in 33.11%, 23.84%, and 15.77% of CN-AML patients, respectively. CEBPA mutations were found at 3 domains: transactivation domain 1 (TAD1) in 10 (6.62%), transactivation domain 2 (TAD2) in 5 (3.31%), and basic leucine zipper domain (bZIP) in 11 (7.82%) patients. Patients with NPM1 mutation had better clinical remission rate (CR) (P=0.003), event-free survival (P=0.0014), and overall survival (OS) (P=0.0017). However, FLT3-ITD and CEBPA mutations did not show any association with CR (P=0.404 and 0.92, respectively). Biallelic CEBPA mutations were found in 12 (7.95%) patients and were associated with better OS (P=0.043). CONCLUSIONS These findings indicate that NPM1 and CEBPA mutations can be precisely used for risk stratification in CN-AML patients.
Collapse
Affiliation(s)
| | | | | | - Jay Singh
- Laboratory Oncology, AIIMSNew Delhi, India
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Shi Y, Xue Y, Wang C, Yu L. Nucleophosmin 1: from its pathogenic role to a tantalizing therapeutic target in acute myeloid leukemia. Hematology 2022; 27:609-619. [PMID: 35621728 DOI: 10.1080/16078454.2022.2067939] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Nucleophosmin 1 (NPM1, also known as B23) is a multifunctional protein involved in a variety of cellular processes, including ribosomal maturation, centrosome replication, maintenance of genomic stability, cell cycle control, and apoptosis. NPM1 is the most commonly mutated gene in adult acute myeloid leukemia (AML) and is present in approximately 40% of all AML cases. The underlying mechanisms of mutant NPM1 (NPM1mut) in leukemogenesis remain unclear. This review summarizes the structure and physiological function of NPM1, mechanisms underlying the pathogenesis of NPM1-mutated AML, and the potential role of NPM1 as a therapeutic target. It is reported that dysfunctional NPM1 might cause AML pathogenesis via its role as a protein chaperone, inhibiting differentiation of leukemia stem cells and regulation of non-coding RNAs. Besides conventional chemotherapies, NPM1 is a promising therapeutic target against AML that warrants further investigation. NPM1-based therapeutic strategies include inducing nucleolar relocalisation of NPM1 mutants, interfering with NPM1 oligomerization, and NPM1 as an immune response target.
Collapse
Affiliation(s)
- Yuye Shi
- Department of Hematology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, People's Republic of China.,Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Yuhao Xue
- Department of Hematology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, People's Republic of China
| | - Chunling Wang
- Department of Hematology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, People's Republic of China.,Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Liang Yu
- Department of Hematology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, People's Republic of China.,Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Xuzhou, People's Republic of China
| |
Collapse
|
9
|
The Role of Nucleophosmin 1 ( NPM1) Mutation in the Diagnosis and Management of Myeloid Neoplasms. LIFE (BASEL, SWITZERLAND) 2022; 12:life12010109. [PMID: 35054502 PMCID: PMC8780493 DOI: 10.3390/life12010109] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 11/17/2022]
Abstract
Nucleophosmin (NPM1) is a multifunctional protein with both proliferative and growth-suppressive roles in the cell. In humans, NPM1 is involved in tumorigenesis via chromosomal translocations, deletions, or mutation. Acute myeloid leukemia (AML) with mutated NPM1, a distinct diagnostic entity by the current WHO Classification of myeloid neoplasm, represents the most common diagnostic subtype in AML and is associated with a favorable prognosis. The persistence of NPM1 mutation in AML at relapse makes this mutation an ideal target for minimal measurable disease (MRD) detection. The clinical implication of this is far-reaching because NPM1-mutated AML is currently classified as being of standard risk, with the best treatment strategy (transplantation versus chemotherapy) yet undefined. Myeloid neoplasms with NPM1 mutations and <20% blasts are characterized by an aggressive clinical course and a rapid progression to AML. The pathological classification of these cases remains controversial. Future studies will determine whether NPM1 gene mutation may be sufficient for diagnosing NPM1-mutated AML independent of the blast count. This review aims to summarize the role of NPM1 in normal cells and in human cancer and discusses its current role in clinical management of AML and related myeloid neoplasms.
Collapse
|
10
|
Histone chaperone Nucleophosmin regulates transcription of key genes involved in oral tumorigenesis. Mol Cell Biol 2021; 42:e0066920. [PMID: 34898280 DOI: 10.1128/mcb.00669-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Nucleophosmin (NPM1) is a multifunctional histone chaperone that can activate acetylation-dependent transcription from chromatin templates in vitro. Acetylation of NPM1 by p300 has been shown to further enhance its transcription activation potential. Moreover, its total and acetylated pools are increased in oral squamous cell carcinoma. However, the role of NPM1 or its acetylated form (AcNPM1) in transcriptional regulation in cells and oral tumorigenesis is not fully elucidated. Using ChIP-seq analyses, we provide the first genome-wide profile of AcNPM1 and show that AcNPM1 is enriched at transcriptional regulatory elements. AcNPM1 co-occupies marks of active transcription at promoters and DNase I hypersensitive sites at enhancers. In addition, using a high-throughput protein interaction profiling approach, we show that NPM1 interacts with RNA Pol II, general transcription factors, mediator subunits, histone acetyltransferase complexes, and chromatin remodelers. NPM1 histone chaperone activity also contributes to its transcription activation potential. Further, NPM1 depletion leads to decreased AcNPM1 occupancy and reduced expression of genes required for proliferative, migratory and invasive potential of oral cancer cells. NPM1 depletion also abrogates the growth of orthotopic tumors in mice. Collectively, these results establish that AcNPM1 functions as a coactivator during during RNA polymerase II-driven transcription and regulates the expression of genes that promote oral tumorigenesis.
Collapse
|
11
|
Molina B, Chavez J, Grainger S. Zebrafish models of acute leukemias: Current models and future directions. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2021; 10:e400. [PMID: 33340278 PMCID: PMC8213871 DOI: 10.1002/wdev.400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/02/2020] [Accepted: 11/09/2020] [Indexed: 12/19/2022]
Abstract
Acute myeloid leukemias (AML) and acute lymphoid leukemias (ALL) are heterogenous diseases encompassing a wide array of genetic mutations with both loss and gain of function phenotypes. Ultimately, these both result in the clonal overgrowth of blast cells in the bone marrow, peripheral blood, and other tissues. As a consequence of this, normal hematopoietic stem cell function is severely hampered. Technologies allowing for the early detection of genetic alterations and understanding of these varied molecular pathologies have helped to advance our treatment regimens toward personalized targeted therapies. In spite of this, both AML and ALL continue to be a major cause of morbidity and mortality worldwide, in part because molecular therapies for the plethora of genetic abnormalities have not been developed. This underscores the current need for better model systems for therapy development. This article reviews the current zebrafish models of AML and ALL and discusses how novel gene editing tools can be implemented to generate better models of acute leukemias. This article is categorized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cells and Disease Technologies > Perturbing Genes and Generating Modified Animals.
Collapse
Affiliation(s)
- Brandon Molina
- Biology Department, San Diego State University, San Diego, California, USA
| | - Jasmine Chavez
- Biology Department, San Diego State University, San Diego, California, USA
| | - Stephanie Grainger
- Biology Department, San Diego State University, San Diego, California, USA
| |
Collapse
|
12
|
Koch J, Lang A, Whongsiri P, Schulz WA, Hoffmann MJ, Greife A. KDM6A mutations promote acute cytoplasmic DNA release, DNA damage response and mitosis defects. BMC Mol Cell Biol 2021; 22:54. [PMID: 34702163 PMCID: PMC8549169 DOI: 10.1186/s12860-021-00394-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 10/13/2021] [Indexed: 12/20/2022] Open
Abstract
Background KDM6A, encoding a histone demethylase, is one of the top ten mutated epigenetic cancer genes. The effect of mutations on its structure and function are however poorly characterized. Methods Database search identified nonsense and missense mutations in the N-terminal TPR motifs and the C-terminal, catalytic JmjC domain, but also in the intrinsically disordered region connecting both these two well-structured domains. KDM6A variants with cancer-derived mutations were generated using site directed mutagenesis and fused to eGFP serving as an all-in-one affinity and fluorescence tag to study demethylase activity by an ELISA-based assay in vitro, apoptosis by FACS, complex assembly by Co-immunoprecipitation and localization by microscopy in urothelial cells and apoptosis by FACS. Results Independent of the mutation and demethylase activity, all KDM6A variants were detectable in the nucleus. Truncated KDM6A variants displayed changes in complex assemblies affecting (1) known interactions with the COMPASS complex component RBBP5 and (2) KDM6A-DNA associated assemblies with the nuclear protein Nucleophosmin. Some KDM6A variants induced a severe cellular phenotype characterized by multiple acute effects on nuclear integrity, namely, release of nuclear DNA into the cytoplasm, increased level of DNA damage indicators RAD51 and p-γH2A.X, and mitosis defects. These damaging effects were correlated with increased cell death. Conclusion These observations reveal novel effects of pathogenic variants pointing at new specific functions of KDM6A variants. The underlying mechanisms and affected pathways have to be investigated in future research to understand how tumor cells cope with and benefit from KDM6A truncations. Supplementary Information The online version contains supplementary material available at 10.1186/s12860-021-00394-2.
Collapse
Affiliation(s)
- J Koch
- Department of Molecular Physical Chemistry, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - A Lang
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - P Whongsiri
- Department of Urology, Medical Faculty, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany.,Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University Bangkog, Bangkok, Thailand
| | - W A Schulz
- Department of Urology, Medical Faculty, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - M J Hoffmann
- Department of Urology, Medical Faculty, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - A Greife
- Department of Molecular Physical Chemistry, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany.
| |
Collapse
|
13
|
Beji S, D'Agostino M, Gambini E, Sileno S, Scopece A, Vinci MC, Milano G, Melillo G, Napolitano M, Pompilio G, Capogrossi MC, Avitabile D, Magenta A. Doxorubicin induces an alarmin-like TLR4-dependent autocrine/paracrine action of Nucleophosmin in human cardiac mesenchymal progenitor cells. BMC Biol 2021; 19:124. [PMID: 34134693 PMCID: PMC8210386 DOI: 10.1186/s12915-021-01058-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 05/28/2021] [Indexed: 12/11/2022] Open
Abstract
Background Doxorubicin (Dox) is an anti-cancer anthracycline drug that causes double-stranded DNA breaks. It is highly effective against several types of tumours; however, it also has adverse effects on regenerative populations of normal cells, such as human cardiac mesenchymal progenitor cells (hCmPCs), and its clinical use is limited by cardiotoxicity. Another known effect of Dox is nucleolar disruption, which triggers the ubiquitously expressed nucleolar phosphoprotein Nucleophosmin (NPM) to be released from the nucleolus into the cell, where it participates in the orchestration of cellular stress responses. NPM has also been observed in the extracellular space in response to different stress stimuli; however, the mechanism behind this and its functional implications are as yet largely unexplored. The aim of this study was to establish whether Dox could elicit NPM secretion in the extracellular space and to elucidate the mechanism of secretion and the effect of extracellular NPM on hCmPCs. Results We found that following the double-strand break formation in hCmPCs caused by Dox, NPM was rapidly secreted in the extracellular space by an active mechanism, in the absence of either apoptosis or necrosis. Extracellular release of NPM was similarly seen in response to ultraviolet radiation (UV). Furthermore, we observed an increase of NPM levels in the plasma of Dox-treated mice; thus, NPM release also occurred in vivo. The treatment of hCmPCs with extracellular recombinant NPM induced a decrease of cell proliferation and a response mediated through the Toll-like receptor (TLR)4. We demonstrated that NPM binds to TLR4, and via TLR4, and nuclear factor kappa B (NFkB) activation/nuclear translocation, exerts proinflammatory functions by inducing IL-6 and COX-2 gene expression. Finally, we found that in hCmPCs, NPM secretion could be driven by an autophagy-dependent unconventional mechanism that requires TLR4, since TLR4 inhibition dramatically reduced Dox-induced secretion. Conclusions We hypothesise that the extracellular release of NPM could be a general response to DNA damage since it can be elicited by either a chemical agent such as Dox or a physical genotoxic stressor such as UV radiation. Following genotoxic stress, NPM acts similarly to an alarmin in hCmPCs, being rapidly secreted and promoting cell cycle arrest and a TLR4/NFκB-dependent inflammatory response. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01058-5.
Collapse
Affiliation(s)
- Sara Beji
- Experimental Immunology Laboratory, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Via Monti di Creta 104, 00167, Rome, Italy
| | - Marco D'Agostino
- Experimental Immunology Laboratory, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Via Monti di Creta 104, 00167, Rome, Italy
| | - Elisa Gambini
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino, IRCCS, Via Carlo Parea 4, 20138, Milan, Italy
| | - Sara Sileno
- Experimental Immunology Laboratory, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Via Monti di Creta 104, 00167, Rome, Italy
| | - Alessandro Scopece
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino, IRCCS, Via Carlo Parea 4, 20138, Milan, Italy
| | - Maria Cristina Vinci
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino, IRCCS, Via Carlo Parea 4, 20138, Milan, Italy
| | - Giuseppina Milano
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino, IRCCS, Via Carlo Parea 4, 20138, Milan, Italy
| | | | | | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino, IRCCS, Via Carlo Parea 4, 20138, Milan, Italy.,Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Maurizio C Capogrossi
- Laboratory of Cardiovascular Science, National Institute on Aging (NIA), National Institutes of Health (NIH), 251 Bayview Blvd, Baltimore, MD, 21224, USA.,Division of Cardiology, Johns Hopkins Bayview Medical Center, Baltimore, MD, USA
| | - Daniele Avitabile
- Idi Farmaceutici S.r.l., Via dei Castelli Romani 83/85, 00071, Pomezia (Rome), Italy.
| | - Alessandra Magenta
- National Research Council of Italy (CNR), Institute of Translational Pharmacology IFT, Via Fosso del Cavaliere 100, 00133, Rome, Italy.
| |
Collapse
|
14
|
López DJ, Rodríguez JA, Bañuelos S. Nucleophosmin, a multifunctional nucleolar organizer with a role in DNA repair. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1868:140532. [PMID: 32853771 DOI: 10.1016/j.bbapap.2020.140532] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/05/2020] [Accepted: 08/07/2020] [Indexed: 12/12/2022]
Abstract
Nucleophosmin (NPM1) is a mostly nucleolar protein with crucial functions in cell growth and homeostasis, including regulation of ribosome biogenesis and stress response. Such multiple activities rely on its ability to interact with nucleic acids and with hundreds of proteins, as well as on a dynamic subcellular distribution. NPM1 is thus regulated by a complex interplay between localization and interactions, further modulated by post-translational modifications. NPM1 is a homopentamer, with globular domains connected by long, intrinsically disordered linkers. This configuration allows NPM1 to engage in liquid-liquid phase separation phenomena, which could underlie a key role in nucleolar organization. Here, we will discuss NPM1 conformational and functional versatility, emphasizing its emerging, and still largely unexplored, role in DNA damage repair. Since NPM1 is altered in a subtype of acute myeloid leukaemia (AML), we will also present ongoing research on the molecular mechanisms underlying its pathogenic role and potential NPM1-targeting therapeutic strategies.
Collapse
Affiliation(s)
- David J López
- Biofisika Institute (UPV/EHU, CSIC) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - José A Rodríguez
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Sonia Bañuelos
- Biofisika Institute (UPV/EHU, CSIC) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Leioa, Spain.
| |
Collapse
|
15
|
Cela I, Di Matteo A, Federici L. Nucleophosmin in Its Interaction with Ligands. Int J Mol Sci 2020; 21:E4885. [PMID: 32664415 PMCID: PMC7402337 DOI: 10.3390/ijms21144885] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/13/2022] Open
Abstract
Nucleophosmin (NPM1) is a mainly nucleolar protein that shuttles between nucleoli, nucleoplasm and cytoplasm to fulfill its many functions. It is a chaperone of both nucleic acids and proteins and plays a role in cell cycle control, centrosome duplication, ribosome maturation and export, as well as the cellular response to a variety of stress stimuli. NPM1 is a hub protein in nucleoli where it contributes to nucleolar organization through heterotypic and homotypic interactions. Furthermore, several alterations, including overexpression, chromosomal translocations and mutations are present in solid and hematological cancers. Recently, novel germline mutations that cause dyskeratosis congenita have also been described. This review focuses on NPM1 interactions and inhibition. Indeed, the list of NPM1 binding partners is ever-growing and, in recent years, many studies contributed to clarifying the structural basis for NPM1 recognition of both nucleic acids and several proteins. Intriguingly, a number of natural and synthetic ligands that interfere with NPM1 interactions have also been reported. The possible role of NPM1 inhibitors in the treatment of multiple cancers and other pathologies is emerging as a new therapeutic strategy.
Collapse
Affiliation(s)
- Ilaria Cela
- Center for Advanced Studies and Technology (CAST), University of Chieti “G. d’Annunzio”, Via Polacchi, 66100 Chieti, Italy;
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti “G. d’Annunzio”, Via dei Vestini 31, 66100 Chieti, Italy
| | - Adele Di Matteo
- Institute of Molecular Biology and Pathology (IBPM) of the CNR, c/o “Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Roma, Italy;
| | - Luca Federici
- Center for Advanced Studies and Technology (CAST), University of Chieti “G. d’Annunzio”, Via Polacchi, 66100 Chieti, Italy;
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti “G. d’Annunzio”, Via dei Vestini 31, 66100 Chieti, Italy
| |
Collapse
|
16
|
Nucleophosmin 1 Mutations in Acute Myeloid Leukemia. Genes (Basel) 2020; 11:genes11060649. [PMID: 32545659 PMCID: PMC7348733 DOI: 10.3390/genes11060649] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [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
|
17
|
Application of a protein domain as chaperone for enhancing biological activity and stability of other proteins. J Biotechnol 2020; 310:68-79. [DOI: 10.1016/j.jbiotec.2020.01.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 01/27/2020] [Accepted: 01/30/2020] [Indexed: 11/21/2022]
|
18
|
Genoveso MJ, Hisaoka M, Komatsu T, Wodrich H, Nagata K, Okuwaki M. Formation of adenovirus DNA replication compartments and viral DNA accumulation sites by host chromatin regulatory proteins including NPM1. FEBS J 2019; 287:205-217. [PMID: 31365788 DOI: 10.1111/febs.15027] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/19/2019] [Accepted: 07/29/2019] [Indexed: 01/08/2023]
Abstract
The adenovirus (Ad) genome is believed to be packaged into the virion by forming a chromatin-like structure. The replicated viral genome is likely to be condensed through binding with viral core proteins before encapsidation. Replicated viral genomes accumulate in the central region of the nucleus, which we termed virus-induced postreplication (ViPR) body. However, the molecular mechanism by which the nuclear structure is reorganized and its functional significance in virus production are currently not understood. In this study, we found that viral packaging protein IVa2, but not capsid proteins, accumulated in the ViPR body. In addition, nucleolar chromatin regulatory proteins, nucleophosmin 1 (NPM1), upstream binding factor, and nucleolin accumulated in the ViPR body in late-stage Ad infection. NPM1 depletion increased the nuclease-resistant viral genome and delayed the ViPR body formation. These results suggested that structural changes in the infected cell nucleus depend on the formation of viral chromatin by host chromatin regulatory proteins. Because NPM1 depletion decreases production of the infectious virion, we propose that host factor-mediated viral chromatin remodeling and concomitant ViPR body formation are prerequisites for efficient encapsidation of Ad chromatin.
Collapse
Affiliation(s)
- Michelle Jane Genoveso
- Ph.D. Program in Humanics, School of Integrative and Global Majors, University of Tsukuba, Japan.,Faculty of Medicine, University of Tsukuba, Japan
| | | | - Tetsuro Komatsu
- CNRS UMR 5234, Fundamental Microbiology and Pathogenicity, Université de Bordeaux, France.,Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Harald Wodrich
- CNRS UMR 5234, Fundamental Microbiology and Pathogenicity, Université de Bordeaux, France
| | | | - Mitsuru Okuwaki
- Faculty of Medicine, University of Tsukuba, Japan.,School of Pharmacy, Kitasato University, Tokyo, Japan
| |
Collapse
|
19
|
Brodská B, Šašinková M, Kuželová K. Nucleophosmin in leukemia: Consequences of anchor loss. Int J Biochem Cell Biol 2019; 111:52-62. [PMID: 31009764 DOI: 10.1016/j.biocel.2019.04.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/17/2019] [Accepted: 04/18/2019] [Indexed: 12/17/2022]
Abstract
Nucleophosmin (NPM), one of the most abundant nucleolar proteins, has crucial functions in ribosome biogenesis, cell cycle control, and DNA-damage repair. In human cells, NPM occurs mainly in oligomers. It functions as a chaperone, undergoes numerous interactions and forms part of many protein complexes. Although NPM role in carcinogenesis is not fully elucidated, a variety of tumor suppressor as well as oncogenic activities were described. NPM is overexpressed, fused with other proteins, or mutated in various tumor types. In the acute myeloid leukemia (AML), characteristic mutations in NPM1 gene, leading to modification of NPM C-terminus, are the most frequent genetic aberration. Although multiple mutation types of NPM are found in AML, they are all characterized by aberrant cytoplasmic localization of the mutated protein. In this review, current knowledge of the structure and function of NPM is presented in relation to its interaction network, in particular to the interaction with other nucleolar proteins and with proteins active in apoptosis. Possible molecular mechanisms of NPM mutation-driven leukemogenesis and NPM therapeutic targeting are discussed. Finally, recent findings concerning the immunogenicity of the mutated NPM and specific immunological features of AML patients with NPM mutation are summarized.
Collapse
Affiliation(s)
- Barbora Brodská
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic
| | - Markéta Šašinková
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic
| | - Kateřina Kuželová
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic.
| |
Collapse
|
20
|
Brunetti L, Gundry MC, Goodell MA. New insights into the biology of acute myeloid leukemia with mutated NPM1. Int J Hematol 2019; 110:150-160. [DOI: 10.1007/s12185-018-02578-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 12/25/2018] [Indexed: 12/20/2022]
|
21
|
Lobaina Y, Perera Y. Implication of B23/NPM1 in Viral Infections, Potential Uses of B23/NPM1 Inhibitors as Antiviral Therapy. Infect Disord Drug Targets 2019; 19:2-16. [PMID: 29589547 DOI: 10.2174/1871526518666180327124412] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/08/2018] [Accepted: 02/12/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND B23/nucleophosmin (B23/NPM1) is an abundant multifunctional protein mainly located in the nucleolus but constantly shuttling between the nucleus and cytosol. As a consequence of its constitutive expression, intracellular dynamics and binding capacities, B23/NPM1 interacts with multiple cellular factors in different cellular compartments, but also with viral proteins from both DNA and RNA viruses. B23/NPM1 influences overall viral replication of viruses like HIV, HBV, HCV, HDV and HPV by playing functional roles in different stages of viral replication including nuclear import, viral genome transcription and assembly, as well as final particle formation. Of note, some virus modify the subcellular localization, stability and/or increases B23/NPM1 expression levels on target cells, probably to foster B23/NPM1 functions in their own replicative cycle. RESULTS This review summarizes current knowledge concerning the interaction of B23/NPM1 with several viral proteins during relevant human infections. The opportunities and challenges of targeting this well-conserved host protein as a potentially new broad antiviral treatment are discussed in detail. Importantly, although initially conceived to treat cancer, a handful of B23/NPM1 inhibitors are currently available to test on viral infection models. CONCLUSION As B23/NPM1 partakes in key steps of viral replication and some viral infections remain as unsolved medical needs, an appealing idea may be the expedite evaluation of B23/NPM1 inhibitors in viral infections. Furthermore, worth to be addressed is if the up-regulation of B23/NPM1 protein levels that follows persistent viral infections may be instrumental to the malignant transformation induced by virus like HBV and HCV.
Collapse
Affiliation(s)
- Yadira Lobaina
- Therapeutic Hepatitis B Vaccine Group, Vaccine Division, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Havana, CP 10600, Cuba
| | - Yasser Perera
- Molecular Oncology Group, Pharmaceuticals Division, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Havana, CP 10600, Cuba
| |
Collapse
|
22
|
Chen S, He H, Wang Y, Liu L, Liu Y, You H, Dong Y, Lyu J. Poor prognosis of nucleophosmin overexpression in solid tumors: a meta-analysis. BMC Cancer 2018; 18:838. [PMID: 30126359 PMCID: PMC6102940 DOI: 10.1186/s12885-018-4718-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 08/02/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Nucleophosmin is a non-ribosomal nucleolar phosphoprotein that is found primarily in the nucleolus region of cell nucleus, plays multiple important roles in tumor processes. Accumulated previous studies have reported a potential value of NPM acted as a biomarker for prognosis in various solid tumors, but the results were more inconsistency. We performed this meta-analysis to precisely evaluate the prognostic significance of NPM in solid tumors. METHODS Clinical data were collected from a comprehensive literature search in PubMed, Web of Science, Embase, and China National Knowledge Infrastructure databases (up to October, 2017). A total of 11 studied with 997 patients were used to assess the association of NPM expression and patients' overall survival (OS). The hazard ratio (HR) or odds ratio (OR) with its 95% confidence intervals (CI) were calculated to estimate the effect. RESULTS The pooled results indicated that higher expression of NPM was observably correlated with poor OS in solid tumor (HR = 1.85, 95% CI: 1.44-2.38, P < 0.001). Furthermore, high expression of NPM was associated with some phenotypes of tumor aggressiveness, such as tumor stage (4 studies, III/IV vs. I/II, OR = 5.21, 95% CI: 2.72-9.56, P < 0.001), differentiation grade (poor vs. well/moderate, OR = 1.82, 95% CI: 1.01-3.27, P = 0.046). CONCLUSION This meta-analysis indicated that NPM may act as a valuable prognosis biomarker and a potential therapeutic target in human solid tumors.
Collapse
Affiliation(s)
- Siying Chen
- Department of Pharmacy, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 of Yanta west road, Xi'an, 710061, Shaanxi, China
| | - Hairong He
- Clinical Research Center, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 of Yanta west road, Xi'an, 710061, Shaanxi, China
| | - Yan Wang
- Department of Pharmacy, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 of Yanta west road, Xi'an, 710061, Shaanxi, China
| | - Leichao Liu
- Department of Pharmacy, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 of Yanta west road, Xi'an, 710061, Shaanxi, China
| | - Yang Liu
- Department of Pharmacy, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 of Yanta west road, Xi'an, 710061, Shaanxi, China
| | - Haisheng You
- Department of Pharmacy, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 of Yanta west road, Xi'an, 710061, Shaanxi, China
| | - Yalin Dong
- Department of Pharmacy, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 of Yanta west road, Xi'an, 710061, Shaanxi, China.
| | - Jun Lyu
- Clinical Research Center, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 of Yanta west road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
23
|
Kaypee S, Sahadevan SA, Sudarshan D, Halder Sinha S, Patil S, Senapati P, Kodaganur GS, Mohiyuddin A, Dasgupta D, Kundu TK. Oligomers of human histone chaperone NPM1 alter p300/KAT3B folding to induce autoacetylation. Biochim Biophys Acta Gen Subj 2018; 1862:1729-1741. [DOI: 10.1016/j.bbagen.2018.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 05/02/2018] [Accepted: 05/04/2018] [Indexed: 11/27/2022]
|
24
|
Derenzini E, Rossi A, Treré D. Treating hematological malignancies with drugs inhibiting ribosome biogenesis: when and why. J Hematol Oncol 2018; 11:75. [PMID: 29855342 PMCID: PMC5984324 DOI: 10.1186/s13045-018-0609-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 04/26/2018] [Indexed: 01/05/2023] Open
Abstract
It is well known that chemotherapy can cure only some cancers in advanced stage, mostly those with an intact p53 pathway. Hematological cancers such as lymphoma and certain forms of leukemia are paradigmatic examples of such scenario. Recent evidence indicates that the efficacy of many of the alkylating and intercalating agents, antimetabolites, topoisomerase, and kinase inhibitors used in cancer therapy is largely due to p53 stabilization and activation consequent to the inhibition of ribosome biogenesis. In this context, innovative drugs specifically hindering ribosome biogenesis showed preclinical activity and are currently in early clinical development in hematological malignancies. The mechanism of p53 stabilization after ribosome biogenesis inhibition is a multistep process, depending on specific factors that can be altered in tumor cells, which can affect the antitumor efficacy of ribosome biogenesis inhibitors (RiBi). In the present review, the basic mechanisms underlying the anticancer activity of RiBi are discussed based on the evidence deriving from available preclinical and clinical studies, with the purpose of defining when and why the treatment with drugs inhibiting ribosomal biogenesis could be highly effective in hematological malignancies.
Collapse
Affiliation(s)
- Enrico Derenzini
- European Institute of Oncology, Via Ripamonti 435, 20141, Milan, Italy.
| | - Alessandra Rossi
- European Institute of Oncology, Via Ripamonti 435, 20141, Milan, Italy
| | - Davide Treré
- DIMES, Università di Bologna, Via Massarenti 9, Bologna, Italy.
| |
Collapse
|
25
|
Abstract
The nucleolus is a prominent subnuclear compartment, where ribosome biosynthesis takes place. Recently, the nucleolus has gained attention for its novel role in the regulation of cellular stress. Nucleolar stress is emerging as a new concept, which is characterized by diverse cellular insult-induced abnormalities in nucleolar structure and function, ultimately leading to activation of p53 or other stress signaling pathways and alterations in cell behavior. Despite a number of comprehensive reviews on this concept, straightforward and clear-cut way criteria for a nucleolar stress state, regarding the factors that elicit this state, the morphological and functional alterations as well as the rationale for p53 activation are still missing. Based on literature of the past two decades, we herein summarize the evolution of the concept and provide hallmarks of nucleolar stress. Along with updated information and thorough discussion of existing confusions in the field, we pay particular attention to the current understanding of the sensing mechanisms, i.e., how stress is integrated by p53. In addition, we propose our own emphasis regarding the role of nucleolar protein NPM1 in the hallmarks of nucleolar stress and sensing mechanisms. Finally, the links of nucleolar stress to human diseases are briefly and selectively introduced.
Collapse
Affiliation(s)
- Kai Yang
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.,Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Jie Yang
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Jing Yi
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| |
Collapse
|
26
|
Kunchala P, Kuravi S, Jensen R, McGuirk J, Balusu R. When the good go bad: Mutant NPM1 in acute myeloid leukemia. Blood Rev 2018; 32:167-183. [DOI: 10.1016/j.blre.2017.11.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 10/19/2017] [Accepted: 11/02/2017] [Indexed: 12/26/2022]
|
27
|
Gollapalli K, Ghantasala S, Atak A, Rapole S, Moiyadi A, Epari S, Srivastava S. Tissue Proteome Analysis of Different Grades of Human Gliomas Provides Major Cues for Glioma Pathogenesis. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2018; 21:275-284. [PMID: 28481733 DOI: 10.1089/omi.2017.0028] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Gliomas are heterogeneous and most commonly occurring brain tumors. Blood-brain barrier restricts the entry of brain tumor proteins into blood stream thus limiting the usage of serum or plasma for proteomic analysis. Our study aimed at understanding the molecular basis of aggressiveness of various grades of brain tumors using isobaric tagging for relative and absolute quantification (iTRAQ) based mass spectrometry. Tissue proteomic analysis of various grades of gliomas was performed using four-plex iTRAQ. We labeled five sets (each set consists of control, grade-II, III, and IV tumor samples) of individual glioma patients using iTRAQ reagents. Significantly altered proteins were subjected to bioinformatics analysis using Database for Annotation, Visualization and Integrated Discovery (DAVID). Various metabolic pathways like glycolysis, TCA-cycle, electron transport chain, lactate metabolism, and blood coagulation pathways were majorly observed to be perturbed in gliomas. Most of the identified proteins involved in redox reactions, protein folding, pre-messenger RNA (mRNA) processing, antiapoptosis, and blood coagulation were found to be upregulated in gliomas. Transcriptomics data of glioblastoma multiforme (GBM), low-grade gliomas (LGGs), and controls were downloaded from The Cancer Genome Atlas (TCGA) data portal and further analyzed using BRB-Array tools. Expression levels of a few significantly altered proteins like lactate dehydrogenase, alpha-1 antitrypsin, fibrinogen alpha chain, nucleophosmin, annexin A5, thioredoxin, ferritin light chain, thymosin beta-4-like protein 3, superoxide dismutase-2, and peroxiredoxin-1 and 6 showed a positive correlation with increasing grade of gliomas thereby offering an insight into molecular basis behind their aggressive nature. Several proteins identified in different grades of gliomas are potential grade-specific markers, and perturbed pathways provide comprehensive overview of molecular cues involved in glioma pathogenesis.
Collapse
Affiliation(s)
- Kishore Gollapalli
- 1 Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay , Mumbai, India
| | - Saicharan Ghantasala
- 1 Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay , Mumbai, India
| | - Apurva Atak
- 1 Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay , Mumbai, India
| | - Srikanth Rapole
- 2 Proteomics Laboratory, National Centre for Cell Science , Pune, India
| | - Aliasgar Moiyadi
- 3 Advanced Center for Treatment Research and Education in Cancer, Tata Memorial Center , Navi Mumbai, India
| | - Sridhar Epari
- 3 Advanced Center for Treatment Research and Education in Cancer, Tata Memorial Center , Navi Mumbai, India
| | - Sanjeeva Srivastava
- 1 Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay , Mumbai, India
| |
Collapse
|
28
|
Luchinat E, Chiarella S, Franceschini M, Di Matteo A, Brunori M, Banci L, Federici L. Identification of a novel nucleophosmin-interaction motif in the tumor suppressor p14arf. FEBS J 2018; 285:832-847. [PMID: 29283500 DOI: 10.1111/febs.14373] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 11/20/2017] [Accepted: 12/20/2017] [Indexed: 12/16/2022]
Abstract
The tumor suppressor p14arf interacts, in response to oncogenic signals, with the p53 E3-ubiquitin ligase HDM2, thereby resulting in p53 stabilization and activation. In addition, it also exerts tumor-suppressive functions in p53-independent contexts. The activities of p14arf are regulated by the nucleolar chaperone nucleophosmin (NPM1), which controls its levels and cellular localization. In acute myeloid leukemia with mutations in the NPM1 gene, mutated NPM1 aberrantly translocates in the cytosol carrying with itself p14arf that is subsequently degraded, thus impairing the p14arf-HDM2-p53 axis. In this work we investigated the complex between these two proteins by means of NMR and other techniques. We identified a novel NPM1-interacting motif in the C-terminal region of p14arf, which corresponds to its predicted nucleolar localization signal. This motif recognizes a specific region of the NPM1 N-terminal domain and, upon binding, the two proteins form soluble high molecular weight complexes. By NMR, we identified critical residues on both proteins involved in the interaction. Collectively, our data provide a structural framework to rationalize the overall assembly of the p14arf-NPM1 supramolecular complexes. A number of p14arf cancer-associated mutations cluster in this motif and their effect on the interaction with NPM1 was also analyzed.
Collapse
Affiliation(s)
- Enrico Luchinat
- CERM, Centro Risonanze Magnetiche, Università di Firenze, Italy.,Dipartimento di Scienze Biomediche, Sperimentali e Cliniche - Università di Firenze, Italy
| | - Sara Chiarella
- Ce.S.I.-MeT Centro di Scienze dell'Invecchiamento e Medicina Traslazionale, Università "G. d'Annunzio" di Chieti, Italy.,Dipartimento di Scienze Mediche, Orali e Biotecnologiche - Università "G. d'Annunzio" di Chieti, Italy
| | - Mimma Franceschini
- Ce.S.I.-MeT Centro di Scienze dell'Invecchiamento e Medicina Traslazionale, Università "G. d'Annunzio" di Chieti, Italy.,Dipartimento di Scienze Mediche, Orali e Biotecnologiche - Università "G. d'Annunzio" di Chieti, Italy
| | - Adele Di Matteo
- Istituto di Biologia e Patologia Molecolari, Consiglio Nazionale delle Ricerche, Roma, Italy
| | - Maurizio Brunori
- Dipartimento di Scienze Biochimiche, "A. Rossi Fanelli" - Sapienza Università di Roma, Italy
| | - Lucia Banci
- CERM, Centro Risonanze Magnetiche, Università di Firenze, Italy.,Dipartimento di Chimica, Università di Firenze, Italy
| | - Luca Federici
- Ce.S.I.-MeT Centro di Scienze dell'Invecchiamento e Medicina Traslazionale, Università "G. d'Annunzio" di Chieti, Italy.,Dipartimento di Scienze Mediche, Orali e Biotecnologiche - Università "G. d'Annunzio" di Chieti, Italy
| |
Collapse
|
29
|
Molecules that target nucleophosmin for cancer treatment: an update. Oncotarget 2018; 7:44821-44840. [PMID: 27058426 PMCID: PMC5190137 DOI: 10.18632/oncotarget.8599] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/28/2016] [Indexed: 11/25/2022] Open
Abstract
Nucleophosmin is a highly and ubiquitously expressed protein, mainly localized in nucleoli but able to shuttle between nucleus and cytoplasm. Nucleophosmin plays crucial roles in ribosome maturation and export, centrosome duplication, cell cycle progression, histone assembly and response to a variety of stress stimuli. Much interest in this protein has arisen in the past ten years, since the discovery of heterozygous mutations in the terminal exon of the NPM1 gene, which are the most frequent genetic alteration in acute myeloid leukemia. Nucleophosmin is also frequently overexpressed in solid tumours and, in many cases, its overexpression correlates with mitotic index and metastatization. Therefore it is considered as a promising target for the treatment of both haematologic and solid malignancies. NPM1 targeting molecules may suppress different functions of the protein, interfere with its subcellular localization, with its oligomerization properties or drive its degradation. In the recent years, several such molecules have been described and here we review what is currently known about them, their interaction with nucleophosmin and the mechanistic basis of their toxicity. Collectively, these molecules exemplify a number of different strategies that can be adopted to target nucleophosmin and we summarize them at the end of the review.
Collapse
|
30
|
Di Matteo A, Franceschini M, Paiardini A, Grottesi A, Chiarella S, Rocchio S, Di Natale C, Marasco D, Vitagliano L, Travaglini-Allocatelli C, Federici L. Structural investigation of nucleophosmin interaction with the tumor suppressor Fbw7γ. Oncogenesis 2017; 6:e379. [PMID: 28920929 PMCID: PMC5623904 DOI: 10.1038/oncsis.2017.78] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 07/17/2017] [Accepted: 07/19/2017] [Indexed: 02/07/2023] Open
Abstract
Nucleophosmin (NPM1) is a multifunctional nucleolar protein implicated in ribogenesis, centrosome duplication, cell cycle control, regulation of DNA repair and apoptotic response to stress stimuli. The majority of these functions are played through the interactions with a variety of protein partners. NPM1 is frequently overexpressed in solid tumors of different histological origin. Furthermore NPM1 is the most frequently mutated protein in acute myeloid leukemia (AML) patients. Mutations map to the C-terminal domain and lead to the aberrant and stable localization of the protein in the cytoplasm of leukemic blasts. Among NPM1 protein partners, a pivotal role is played by the tumor suppressor Fbw7γ, an E3-ubiquitin ligase that degrades oncoproteins like c-MYC, cyclin E, Notch and c-jun. In AML with NPM1 mutations, Fbw7γ is degraded following its abnormal cytosolic delocalization by mutated NPM1. This mechanism also applies to other tumor suppressors and it has been suggested that it may play a key role in leukemogenesis. Here we analyse the interaction between NPM1 and Fbw7γ, by identifying the protein surfaces implicated in recognition and key aminoacids involved. Based on the results of computational methods, we propose a structural model for the interaction, which is substantiated by experimental findings on several site-directed mutants. We also extend the analysis to two other NPM1 partners (HIV Tat and CENP-W) and conclude that NPM1 uses the same molecular surface as a platform for recognizing different protein partners. We suggest that this region of NPM1 may be targeted for cancer treatment.
Collapse
Affiliation(s)
- A Di Matteo
- Istituto di Biologia e Patologia Molecolari - Consiglio Nazionale delle ricerche, Roma, Italy
| | - M Franceschini
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, Chieti, Italy.,CeSI-Met - Università di Chieti-Pescara 'G d'Annunzio', Chieti, Italy
| | - A Paiardini
- Dipartimento di Biologia e Biotecnologie 'C Darwin' - Sapienza Università di Roma, Roma, Italy
| | - A Grottesi
- CINECA Consorzio Interuniversitario, Sede di Roma, Roma, Italy
| | - S Chiarella
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, Chieti, Italy.,CeSI-Met - Università di Chieti-Pescara 'G d'Annunzio', Chieti, Italy
| | - S Rocchio
- Dipartimento di Scienze Biochimiche 'A Rossi Fanelli' - Sapienza Università di Roma, Roma, Italy
| | - C Di Natale
- Dipartimento di Farmacia,- Università di Napoli 'Federico II', Napoli, Italy
| | - D Marasco
- Dipartimento di Farmacia,- Università di Napoli 'Federico II', Napoli, Italy
| | - L Vitagliano
- Istituto di Biostrutture e Bioimmagini - Consiglio Nazionale delle Ricerche, Napoli, Italy
| | - C Travaglini-Allocatelli
- Dipartimento di Scienze Biochimiche 'A Rossi Fanelli' - Sapienza Università di Roma, Roma, Italy
| | - L Federici
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, Chieti, Italy.,CeSI-Met - Università di Chieti-Pescara 'G d'Annunzio', Chieti, Italy
| |
Collapse
|
31
|
Hanley ML, Yoo TY, Sonnett M, Needleman DJ, Mitchison TJ. Chromosomal passenger complex hydrodynamics suggests chaperoning of the inactive state by nucleoplasmin/nucleophosmin. Mol Biol Cell 2017; 28:1444-1456. [PMID: 28404751 PMCID: PMC5449145 DOI: 10.1091/mbc.e16-12-0860] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/27/2017] [Accepted: 04/04/2017] [Indexed: 01/30/2023] Open
Abstract
The chromosomal passenger complex (CPC) is a conserved, essential regulator of cell division. As such, significant anti-cancer drug development efforts have been focused on targeting it, most notably by inhibiting its AURKB kinase subunit. The CPC is activated by AURKB-catalyzed autophosphorylation on multiple subunits, but how this regulates CPC interactions with other mitotic proteins remains unclear. We investigated the hydrodynamic behavior of the CPC in Xenopus laevis egg cytosol using sucrose gradient sedimentation and in HeLa cells using fluorescence correlation spectroscopy. We found that autophosphorylation of the CPC decreases its sedimentation coefficient in egg cytosol and increases its diffusion coefficient in live cells, indicating a decrease in mass. Using immunoprecipitation coupled with mass spectrometry and immunoblots, we discovered that inactive, unphosphorylated CPC interacts with nucleophosmin/nucleoplasmin proteins, which are known to oligomerize into pentamers and decamers. Autophosphorylation of the CPC causes it to dissociate from nucleophosmin/nucleoplasmin. We propose that nucleophosmin/nucleoplasmin complexes serve as chaperones that negatively regulate the CPC and/or stabilize its inactive form, preventing CPC autophosphorylation and recruitment to chromatin and microtubules in mitosis.
Collapse
Affiliation(s)
- Mariah L Hanley
- Department of Systems Biology, Harvard Medical School, Boston, MA 02114-5701.,Department of Chemistry, Harvard University, Cambridge, MA 02138-2902
| | - Tae Yeon Yoo
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138-2902
| | - Matthew Sonnett
- Department of Systems Biology, Harvard Medical School, Boston, MA 02114-5701
| | - Daniel J Needleman
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138-2902.,Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138-2902
| | - Timothy J Mitchison
- Department of Systems Biology, Harvard Medical School, Boston, MA 02114-5701
| |
Collapse
|
32
|
Cheung DHC, Ho ST, Lau KF, Jin R, Wang YN, Kung HF, Huang JJ, Shaw PC. Nucleophosmin Interacts with PIN2/TERF1-interacting Telomerase Inhibitor 1 (PinX1) and Attenuates the PinX1 Inhibition on Telomerase Activity. Sci Rep 2017; 7:43650. [PMID: 28255170 PMCID: PMC5334639 DOI: 10.1038/srep43650] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 01/27/2017] [Indexed: 11/26/2022] Open
Abstract
Telomerase activation and telomere maintenance are critical for cellular immortalization and transformation. PIN2/TERF1-interacting telomerase inhibitor 1 (PinX1) is a telomerase regulator and the aberrant expression of PinX1 causes telomere shortening. Identifying PinX1-interacting proteins is important for understanding telomere maintenance. We found that PinX1 directly interacts with nucleophosmin (NPM), a protein that has been shown to positively correlate with telomerase activity. We further showed that PinX1 acts as a linker in the association between NPM and hTERT, the catalytic subunit of telomerase. Additionally, the recruitment of NPM by PinX1 to the telomerase complex could partially attenuate the PinX1-mediated inhibition on telomerase activity. Taken together, our data reveal a novel mechanism that regulates telomerase activation through the interaction between NPM, PinX1 and the telomerase complex.
Collapse
Affiliation(s)
- Derek Hang-Cheong Cheung
- Centre for Protein Science and Crystallography, School of Life Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Sai-Tim Ho
- Centre for Protein Science and Crystallography, School of Life Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Kwok-Fai Lau
- Centre for Protein Science and Crystallography, School of Life Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Rui Jin
- Laboratory of Tumor and Molecular Biology, Beijing Institute of Biotechnology, Beijing, China
| | - Ya-Nan Wang
- Laboratory of Tumor and Molecular Biology, Beijing Institute of Biotechnology, Beijing, China
| | - Hsiang-Fu Kung
- Stanley Ho Center for Emerging Infectious Diseases, Li Ka-Shing Medical Institute, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Jun-Jian Huang
- Laboratory of Tumor and Molecular Biology, Beijing Institute of Biotechnology, Beijing, China
| | - Pang-Chui Shaw
- Centre for Protein Science and Crystallography, School of Life Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| |
Collapse
|
33
|
Shi D, Shi H, Sun D, Chen J, Zhang X, Wang X, Zhang J, Ji Z, Liu J, Cao L, Zhu X, Yuan J, Dong H, Wang X, Chang T, Liu Y, Feng L. Nucleocapsid Interacts with NPM1 and Protects it from Proteolytic Cleavage, Enhancing Cell Survival, and is Involved in PEDV Growth. Sci Rep 2017; 7:39700. [PMID: 28045037 PMCID: PMC5206633 DOI: 10.1038/srep39700] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 11/22/2016] [Indexed: 12/24/2022] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) replicates in the cytoplasm of infected cells, but its nucleocapsid (N) protein localizes specifically to the nucleolus. The mechanism of nuclear translocation, and whether N protein associates with particular nucleolar components, is unknown. In this study, we confirm that a nucleolar phosphoprotein nucleophosmin (NPM1) interacts and co-localizes with the N protein in the nucleolus. In vitro binding studies indicated that aa 148–294 of N and aa 118–188 of NPM1 were required for binding. Interestingly, N protein importation into the nucleolus is independent of the ability of NPM1 to shuttle between the nucleus and the cytoplasm. Furthermore, overexpression of NPM1 promoted PEDV growth, while knockdown of NPM1 suppressed PEDV growth. In addition, binding of N protein to NPM1 protects it from proteolytic degradation by caspase-3, leading to increased cell survival. Taken together, our studies demonstrate a specific interaction of the N protein with the host cell protein NPM1 in the nucleolus. The results suggest potential linkages among viral strategies for the regulation of cell survival activities, possibly through an interaction of N protein with NPM1 which prevents its proteolytic cleavage and enhances cell survival, thus ultimately promoting the replication of PEDV.
Collapse
Affiliation(s)
- Da Shi
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin 150069, P. R. China
| | - Hongyan Shi
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin 150069, P. R. China
| | - Dongbo Sun
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 2 Xinyang Road, Sartu District, Daqing 163319, P. R. China
| | - Jianfei Chen
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin 150069, P. R. China
| | - Xin Zhang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin 150069, P. R. China
| | - Xiaobo Wang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin 150069, P. R. China
| | - Jialin Zhang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin 150069, P. R. China
| | - Zhaoyang Ji
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin 150069, P. R. China
| | - Jianbo Liu
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin 150069, P. R. China
| | - Liyan Cao
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin 150069, P. R. China
| | - Xiangdong Zhu
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin 150069, P. R. China
| | - Jing Yuan
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin 150069, P. R. China
| | - Hui Dong
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin 150069, P. R. China
| | - Xin Wang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin 150069, P. R. China
| | - Tiecheng Chang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin 150069, P. R. China
| | - Ye Liu
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin 150069, P. R. China
| | - Li Feng
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin 150069, P. R. China
| |
Collapse
|
34
|
Lin J, Hisaoka M, Nagata K, Okuwaki M. Functional characterization and efficient detection of Nucleophosmin/NPM1 oligomers. Biochem Biophys Res Commun 2016; 480:702-708. [PMID: 27983985 DOI: 10.1016/j.bbrc.2016.10.125] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 10/27/2016] [Indexed: 11/16/2022]
Abstract
NPM1/nucleophosmin is a multifunctional and oligomeric phosphoprotein. A number of observations have suggested that changes in the oligomer formation of NPM1 could influence its biological functions, especially its oncogenic functions. To understand the functional meaning of oligomerization of NPM1/nucleophosmin, we have established a novel method to monitor protein oligomerization in cells. We utilized the split synthetic Renilla luciferase protein fragment-assisted complementation (SRL-PFAC) bioluminescence activity and observed the change of NPM1 oligomer levels under various cell culture conditions. Our study provides a method for systematic characterization of NPM1 oligomer formation changes and for screening inhibitors of NPM1 oligomerization.
Collapse
Affiliation(s)
- Jianhuang Lin
- PhD Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, 305-8575, Japan; Department of Infection Biology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, 305-8575, Japan
| | - Miharu Hisaoka
- Department of Infection Biology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, 305-8575, Japan
| | - Kyosuke Nagata
- Department of Infection Biology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, 305-8575, Japan
| | - Mitsuru Okuwaki
- PhD Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, 305-8575, Japan; Department of Infection Biology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, 305-8575, Japan.
| |
Collapse
|
35
|
Scott DD, Oeffinger M. Nucleolin and nucleophosmin: nucleolar proteins with multiple functions in DNA repair. Biochem Cell Biol 2016; 94:419-432. [PMID: 27673355 DOI: 10.1139/bcb-2016-0068] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The nucleolus represents a highly multifunctional intranuclear organelle in which, in addition to the canonical ribosome assembly, numerous processes such as transcription, DNA repair and replication, the cell cycle, and apoptosis are coordinated. The nucleolus is further a key hub in the sensing of cellular stress and undergoes major structural and compositional changes in response to cellular perturbations. Numerous nucleolar proteins have been identified that, upon sensing nucleolar stress, deploy additional, non-ribosomal roles in the regulation of varied cell processes including cell cycle arrest, arrest of DNA replication, induction of DNA repair, and apoptosis, among others. The highly abundant proteins nucleophosmin (NPM1) and nucleolin (NCL) are two such factors that transit to the nucleoplasm in response to stress, and participate directly in the repair of numerous different DNA damages. This review discusses the contributions made by NCL and (or) NPM1 to the different DNA repair pathways employed by mammalian cells to repair DNA insults, and examines the implications of such activities for the regulation, pathogenesis, and therapeutic targeting of NPM1 and NCL.
Collapse
Affiliation(s)
- Daniel D Scott
- a Laboratory of RNP Biochemistry, Institut de recherches cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada
- b Division of Experimental Medicine, Faculty of Medicine, McGill University, Montréal, QC H3A 2A3, Canada
| | - Marlene Oeffinger
- a Laboratory of RNP Biochemistry, Institut de recherches cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada
- b Division of Experimental Medicine, Faculty of Medicine, McGill University, Montréal, QC H3A 2A3, Canada
- c Département de biochimie et médecine moléculaire, Faculté de Médecine, Université de Montréal, QC H3T 1J4, Canada
| |
Collapse
|
36
|
Woerner AC, Frottin F, Hornburg D, Feng LR, Meissner F, Patra M, Tatzelt J, Mann M, Winklhofer KF, Hartl FU, Hipp MS. Cytoplasmic protein aggregates interfere with nucleocytoplasmic transport of protein and RNA. Science 2015; 351:173-6. [DOI: 10.1126/science.aad2033] [Citation(s) in RCA: 293] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 11/11/2015] [Indexed: 12/12/2022]
|
37
|
Biophysical Characterization of Nucleophosmin Interactions with Human Immunodeficiency Virus Rev and Herpes Simplex Virus US11. PLoS One 2015; 10:e0143634. [PMID: 26624888 PMCID: PMC4704560 DOI: 10.1371/journal.pone.0143634] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 11/06/2015] [Indexed: 02/07/2023] Open
Abstract
Nucleophosmin (NPM1, also known as B23, numatrin or NO38) is a pentameric RNA-binding protein with RNA and protein chaperon functions. NPM1 has increasingly emerged as a potential cellular factor that directly associates with viral proteins; however, the significance of these interactions in each case is still not clear. In this study, we have investigated the physical interaction of NPM1 with both human immunodeficiency virus type 1 (HIV-1) Rev and Herpes Simplex virus type 1 (HSV-1) US11, two functionally homologous proteins. Both viral proteins show, in mechanistically different modes, high affinity for a binding site on the N-terminal oligomerization domain of NPM1. Rev, additionally, exhibits low-affinity for the central histone-binding domain of NPM1. We also showed that the proapoptotic cyclic peptide CIGB-300 specifically binds to NPM1 oligomerization domain and blocks its association with Rev and US11. Moreover, HIV-1 virus production was significantly reduced in the cells treated with CIGB-300. Results of this study suggest that targeting NPM1 may represent a useful approach for antiviral intervention.
Collapse
|
38
|
Ling H, Hanashiro K, Luong TH, Benavides L, Fukasawa K. Functional relationship among PLK2, PLK4 and ROCK2 to induce centrosome amplification. Cell Cycle 2015; 14:544-53. [PMID: 25590559 DOI: 10.4161/15384101.2014.989121] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The presence of more than 2 centrosomes (centrosome amplification) leads to defective mitosis and chromosome segregation errors, is frequently found in a variety of cancer types, and believed to be the major cause of chromosome instability. One mechanism for generation of amplified centrosomes is over-duplication of centrosomes in a single cell cycle, which is expected to occur when cells are temporarily arrested. There are a growing number of kinases that are critical for induction and promotion of centrosome amplification in the cell cycle-arrested cells, including Rho-associated kinase (ROCK2), Polo-like kinase 2 (PLK2) and PLK4. Here, we tested whether these kinases induce centrosome amplification in a linear pathway or parallel pathways. We first confirmed that ROCK2, PLK2 and PLK4 are all essential for centrosomes to re-duplicate in the cells arrested by exposure to DNA synthesis inhibitor. Using the centrosome amplification rescue assay, we found that PLK2 indirectly activates ROCK2 via phosphorylating nucleophosmin (NPM), and PLK4 functions downstream of ROCK2 to drive centrosome amplification in the arrested cells.
Collapse
Key Words
- Aph, aphidicolin
- B23
- CDK2, Cyclin-dependent kinase 2
- DN, dominant-negative
- KD, kinase-dead
- MEFs, mouse embryonic fibroblasts
- NPM
- NPM, Nucleophosmin
- PLK2
- PLK2, Polo-like kinase 2
- PLK4
- PLK4, Polo-like kinase 4
- Puro, puromycin
- ROCK2
- ROCK2, Rho-(associated) kinase
- centrosome
- nucleophosmin
- polo-like kinase
- wt, wild type
Collapse
Affiliation(s)
- Hongbo Ling
- a Department of Molecular Oncology ; H. Lee Moffitt Cancer Center & Research Institute ; Tampa , FL USA
| | | | | | | | | |
Collapse
|
39
|
Kim JY, Cho YE, An YM, Kim SH, Lee YG, Park JH, Lee S. GLTSCR2 is an upstream negative regulator of nucleophosmin in cervical cancer. J Cell Mol Med 2015; 19:1245-52. [PMID: 25818168 PMCID: PMC4459840 DOI: 10.1111/jcmm.12474] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 09/24/2014] [Indexed: 11/29/2022] Open
Abstract
Nucleophosmin (NPM)/B23, a multifunctional nucleolar phosphoprotein, plays an important role in ribosome biogenesis, cell cycle regulation, apoptosis and cancer pathogenesis. The role of NPM in cells is determined by several factors, including total expression level, oligomerization or phosphorylation status, and subcellular localization. In the nucleolus, NPM participates in rRNA maturation to enhance ribosomal biogenesis. Consistent with this finding, NPM expression is increased in rapidly proliferating cells and many types of human cancers. In response to ribosomal stress, NPM is redistributed to the nucleoplasm, where it inactivates mouse double minute 2 homologue to stabilize p53 and inhibit cell cycle progression. These observations indicate that nucleolus-nucleoplasmic mobilization of NPM is one of the key molecular mechanisms that determine the role of NPM within the cell. However, the regulatory molecule(s) that control(s) NPM stability and subcellular localization, crucial to the pluripotency of intercellular NPM, remain(s) unidentified. In this study, we showed that nucleolar protein GLTSCR2/Pict-1 induced nucleoplasmic translocation and enhanced the degradation of NPM via the proteasomal polyubiquitination pathway. In addition, we showed that GLTSCR2 expression decreased the transforming activity of cells mediated by NPM and that the expression of NPM is reciprocally related to that of GLTSCR2 in cervical cancer tissue. In this study, we demonstrated that GLTSCR2 is an upstream negative regulator of NPM.
Collapse
Affiliation(s)
- Jee-Youn Kim
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Young-Eun Cho
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Yong-Min An
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Sang-Hoon Kim
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Yong-Gwan Lee
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Jae-Hoon Park
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Sun Lee
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul, Korea
| |
Collapse
|
40
|
Mukherjee H, Chan KP, Andresen V, Hanley ML, Gjertsen BT, Myers AG. Interactions of the natural product (+)-avrainvillamide with nucleophosmin and exportin-1 Mediate the cellular localization of nucleophosmin and its AML-associated mutants. ACS Chem Biol 2015; 10:855-63. [PMID: 25531824 PMCID: PMC4652655 DOI: 10.1021/cb500872g] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nucleophosmin (NPM1) is a multifunctional phosphoprotein localized predominantly within the nucleoli of eukaryotic cells. Mutations within its C-terminal domain are frequently observed in patients with acute myeloid leukemia (AML), are thought to play a key role in the initiation of the disease, and result in aberrant, cytoplasmic localization of the mutant protein. We have previously shown that the electrophilic antiproliferative natural product (+)-avrainvillamide (1) binds to proteins, including nucleophosmin, by S-alkylation of cysteine residues. Here, we report that avrainvillamide restores nucleolar localization of certain AML-associated mutant forms of NPM1 and provide evidence that this relocalization is mediated by interactions of avrainvillamide with mutant NPM1 and exportin-1 (Crm1). Immunofluorescence and mass spectrometric experiments employing a series of different NPM1 constructs suggest that a specific interaction between avrainvillamide and Cys275 of certain NPM1 mutants mediates the relocalization of these proteins to the nucleolus. Avrainvillamide treatment is also shown to inhibit nuclear export of Crm1 cargo proteins, including AML-associated NPM1 mutants. We also observe that avrainvillamide treatment displaces Thr199-phosphorylated NPM1 from duplicated centrosomes, leads to an accumulation of supernumerary centrosomes, and inhibits dephosphorylation of Thr199-phosphorylated NPM1 by protein phosphatase 1. Avrainvillamide is the first small molecule reported to relocalize specific cytoplasmic AML-associated NPM1 mutants to the nucleolus, providing an important demonstration of principle that small molecule induction of a wild-type NPM1 localization phenotype is feasible in certain human cancer cells.
Collapse
Affiliation(s)
- Herschel Mukherjee
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States
| | - Kok-Ping Chan
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States
| | - Vibeke Andresen
- Centre for Cancer Biomarkers, CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Mariah L. Hanley
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States
| | - Bjørn Tore Gjertsen
- Centre for Cancer Biomarkers, CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
| | - Andrew G. Myers
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States
| |
Collapse
|
41
|
Abstract
Human rhinoviruses (HRV) are the major etiological agents of the common cold and asthma exacerbations, with significant worldwide health and economic impact. Although large-scale population vaccination has proved successful in limiting or even eradicating many viruses, the more than 100 distinct serotypes mean that conventional vaccination is not a feasible strategy to combat HRV. An alternative strategy is to target conserved viral proteins such as the HRV proteases, 2A(pro) and 3C(pro), the focus of this review. Necessary for host cell shutoff, virus replication, and pathogenesis, 2A(pro) and 3C(pro) are clearly viable drug targets, and indeed, 3C(pro) has been successfully targeted for treating the common cold in experimental infection. 2A(pro) and 3C(pro) are crucial for virus replication due to their role in polyprotein processing as well as cleavage of key cellular proteins to inhibit cellular transcription and translation. Intriguingly, the action of the HRV proteases also disrupts nucleocytoplasmic trafficking, contributing to HRV cytopathic effects. Improved understanding of the protease-cell interactions should enable new therapeutic approaches to be identified for drug development.
Collapse
Affiliation(s)
- Lora M Jensen
- Faculty of Education, Science, Technology and Mathematics, Centre for Research in Therapeutic Solutions, University of Canberra, 1 Kirinari Street, Bruce, Canberra, ACT, 2601, Australia
| | | | | | | |
Collapse
|
42
|
Dynamic conformations of nucleophosmin (NPM1) at a key monomer-monomer interface affect oligomer stability and interactions with granzyme B. PLoS One 2014; 9:e115062. [PMID: 25490769 PMCID: PMC4260957 DOI: 10.1371/journal.pone.0115062] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 11/05/2014] [Indexed: 01/02/2023] Open
Abstract
Nucleophosmin (NPM1) is an abundant, nucleolar tumor antigen with important roles in cell proliferation and putative contributions to oncogenesis. Wild-type NPM1 forms pentameric oligomers through interactions at the amino-terminal core domain. A truncated form of NPM1 found in some hepatocellular carcinoma tissue formed an unusually stable oligomer and showed increased susceptibility to cleavage by granzyme B. Initiation of translation at the seventh methionine generated a protein (M7-NPM) that shared all these properties. We used deuterium exchange mass spectrometry (DXMS) to perform a detailed structural analysis of wild-type NPM1 and M7-NPM, and found dynamic conformational shifts or local “unfolding” at a specific monomer-monomer interface which included the β-hairpin “latch.” We tested the importance of interactions at the β-hairpin “latch” by replacing a conserved tyrosine in the middle of the β-hairpin loop with glutamic acid, generating Y67E-NPM. Y67E-NPM did not form stable oligomers and further, prevented wild-type NPM1 oligomerization in a dominant-negative fashion, supporting the critical role of the β-hairpin “latch” in monomer-monomer interactions. Also, we show preferential cleavage by granzyme B at one of two available aspartates (either D161 or D122) in M7-NPM and Y67E-NPM, whereas wild-type NPM1 was cleaved at both sites. Thus, we observed a correlation between the propensity to form oligomers and granzyme B cleavage site selection in nucleophosmin proteins, suggesting that a small change at an important monomer-monomer interface can affect conformational shifts and impact protein-protein interactions.
Collapse
|
43
|
Abstract
The intracellular location and regulation of proteins within each cell is critically important and is typically deregulated in disease especially cancer. The clinical hypothesis for inhibiting the nucleo-cytoplasmic transport is based on the dependence of certain key proteins within malignant cells. This includes a host of well-characterized tumor suppressor and oncoproteins that require specific localization for their function. This aberrant localization of tumour suppressors and oncoproteins results in their their respective inactivation or over-activation. This incorrect localization occurs actively via the nuclear pore complex that spans the nuclear envelope and is mediated by transport receptors. Accordingly, given the significant need for novel, specific disease treatments, the nuclear envelope and the nuclear transport machinery have emerged as a rational therapeutic target in oncology to restore physiological nucleus/cytoplasmic homeostasis. Recent evidence suggests that this approach might be of substantial therapeutic use. This review summarizes the mechanisms of nucleo-cytoplasmic transport, its role in cancer biology and the therapeutic potential of targeting this critical cellular process.
Collapse
Affiliation(s)
- Richard Hill
- Regenerative Medicine Program, Departamento de Ciências Biomédicas e Medicina, Universidade do Algarve, Portugal
| | | | | | | |
Collapse
|
44
|
Lührig S, Siamishi I, Tesmer-Wolf M, Zechner U, Engel W, Nolte J. Lrrc34, a novel nucleolar protein, interacts with npm1 and ncl and has an impact on pluripotent stem cells. Stem Cells Dev 2014; 23:2862-74. [PMID: 24991885 PMCID: PMC4236065 DOI: 10.1089/scd.2013.0470] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 07/02/2014] [Indexed: 11/13/2022] Open
Abstract
The gene Lrrc34 (leucine rich repeat containing 34) is highly expressed in pluripotent stem cells and its expression is strongly downregulated upon differentiation. These results let us to suggest a role for Lrrc34 in the regulation and maintenance of pluripotency. Expression analyses revealed that Lrrc34 is predominantly expressed in pluripotent stem cells and has an impact on the expression of known pluripotency genes, such as Oct4. Methylation studies of the Lrrc34 promoter showed a hypomethylation in undifferentiated stem cells and chromatin immunoprecipitation-quantitative polymerase chain reaction analyses of histone modifications revealed an enrichment of activating histone modifications on the Lrrc34 promoter region. Further, we could verify the nucleolus-the place of ribosome biogenesis-as the major subcellular localization of the LRRC34 protein. We have verified the interaction of LRRC34 with two major nucleolar proteins, Nucleophosmin and Nucleolin, by two independent methods, suggesting a role for Lrrc34 in ribosome biogenesis of pluripotent stem cells. In conclusion, LRRC34 is a novel nucleolar protein that is predominantly expressed in pluripotent stem cells. Its altered expression has an impact on pluripotency-regulating genes and it interacts with proteins known to be involved in ribosome biogenesis. Therefore we suggest a role for Lrrc34 in ribosome biogenesis of pluripotent stem cells.
Collapse
Affiliation(s)
- Sandra Lührig
- Institute of Human Genetics, University of Göttingen, Göttingen, Germany
| | - Iliana Siamishi
- Institute of Human Genetics, University of Göttingen, Göttingen, Germany
| | | | - Ulrich Zechner
- Institute of Human Genetics, University of Mainz, Mainz, Germany
| | - Wolfgang Engel
- Institute of Human Genetics, University of Göttingen, Göttingen, Germany
| | - Jessica Nolte
- Institute of Human Genetics, University of Göttingen, Göttingen, Germany
| |
Collapse
|
45
|
Ren Z, Aerts JL, Pen JJ, Heirman C, Breckpot K, De Grève J. Phosphorylated STAT3 physically interacts with NPM and transcriptionally enhances its expression in cancer. Oncogene 2014; 34:1650-7. [PMID: 24793791 DOI: 10.1038/onc.2014.109] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 03/04/2014] [Accepted: 03/17/2014] [Indexed: 12/15/2022]
Abstract
The signal transducer and activator of transcription 3 (STAT3) can be activated by the tyrosine kinase domain of the chimeric protein nucleophosmin/anaplastic lymphoma kinase (NPM/ALK), and has a pivotal role in mediating NPM/ALK-related malignant cell transformation. Although the role of STAT3 and wild-type NPM in oncogenesis has been extensively investigated, the relationship between both molecules in cancer remains poorly understood. In the present study, we first demonstrate that STAT3 phosphorylation at tyrosine 705 is accompanied by a concomitant increase in the expression level of NPM. Nuclear co-translocation of phosphorylated STAT3 with NPM can be triggered by interferon-alpha (IFN-α) stimulation of Jurkat cells and phosphorylated STAT3 co-localizes with NPM in cancer cells showing constitutive STAT3 activation. We further demonstrate that STAT3 phosphorylation can transcriptionally mediate NPM upregulation in IFN-α-stimulated Jurkat cells and is responsible for maintaining its expression in cancer cells showing constitutive STAT3 activation. Inhibition of STAT3 phosphorylation or knockdown of NPM expression abrogates their simultaneous transnuclear movements. Finally, we found evidence for a physical interaction between NPM and STAT3 in conditions of STAT3 activation. In conclusion, NPM is a downstream effector of the STAT3 signaling, and can facilitate the nuclear entry of phosphorylated STAT3. These observations might open novel opportunities for targeting the STAT3 pathway in cancer.
Collapse
Affiliation(s)
- Z Ren
- 1] Laboratory of Medical and Molecular Oncology (LMMO), Department of Medical Oncology, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium [2] Department of General Surgery, The first People's Hospital of Shanghai, Shanghai Jiaotong University, Shanghai, China [3] Department of Medical Oncology, Oncologisch Centrum of the Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - J L Aerts
- Department of Immunology-Physiology, Vrije Universiteit Brussel, Brussels, Belgium
| | - J J Pen
- 1] Department of Medical Oncology, Oncologisch Centrum of the Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium [2] Department of Immunology-Physiology, Vrije Universiteit Brussel, Brussels, Belgium
| | - C Heirman
- Department of Immunology-Physiology, Vrije Universiteit Brussel, Brussels, Belgium
| | - K Breckpot
- Department of Immunology-Physiology, Vrije Universiteit Brussel, Brussels, Belgium
| | - J De Grève
- 1] Laboratory of Medical and Molecular Oncology (LMMO), Department of Medical Oncology, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium [2] Department of Medical Oncology, Oncologisch Centrum of the Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
46
|
Maggi LB, Winkeler CL, Miceli AP, Apicelli AJ, Brady SN, Kuchenreuther MJ, Weber JD. ARF tumor suppression in the nucleolus. Biochim Biophys Acta Mol Basis Dis 2014; 1842:831-9. [PMID: 24525025 DOI: 10.1016/j.bbadis.2014.01.016] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 01/27/2014] [Accepted: 01/28/2014] [Indexed: 02/06/2023]
Abstract
Since its discovery close to twenty years ago, the ARF tumor suppressor has played a pivotal role in the field of cancer biology. Elucidating ARF's basal physiological function in the cell has been the focal interest of numerous laboratories throughout the world for many years. Our current understanding of ARF is constantly evolving to include novel frameworks for conceptualizing the regulation of this critical tumor suppressor. As a result of this complexity, there is great need to broaden our understanding of the intricacies governing the biology of the ARF tumor suppressor. The ARF tumor suppressor is a key sensor of signals that instruct a cell to grow and proliferate and is appropriately localized in nucleoli to limit these processes. This article is part of a Special Issue entitled: Role of the Nucleolus in Human Disease.
Collapse
Affiliation(s)
- Leonard B Maggi
- BRIGHT Institute, Department of Internal Medicine, Division of Molecular Oncology, Siteman Cancer Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Crystal L Winkeler
- BRIGHT Institute, Department of Internal Medicine, Division of Molecular Oncology, Siteman Cancer Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Alexander P Miceli
- BRIGHT Institute, Department of Internal Medicine, Division of Molecular Oncology, Siteman Cancer Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Anthony J Apicelli
- BRIGHT Institute, Department of Internal Medicine, Division of Molecular Oncology, Siteman Cancer Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Suzanne N Brady
- BRIGHT Institute, Department of Internal Medicine, Division of Molecular Oncology, Siteman Cancer Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Michael J Kuchenreuther
- BRIGHT Institute, Department of Internal Medicine, Division of Molecular Oncology, Siteman Cancer Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jason D Weber
- BRIGHT Institute, Department of Internal Medicine, Division of Molecular Oncology, Siteman Cancer Center, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
47
|
Leal MF, Mazzotti TKF, Calcagno DQ, Cirilo PDR, Martinez MC, Demachki S, Assumpção PP, Chammas R, Burbano RR, Smith MC. Deregulated expression of Nucleophosmin 1 in gastric cancer and its clinicopathological implications. BMC Gastroenterol 2014; 14:9. [PMID: 24410879 PMCID: PMC3893589 DOI: 10.1186/1471-230x-14-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 12/31/2013] [Indexed: 11/19/2022] Open
Abstract
Background The process of gastric carcinogenesis still remains to be elucidated. The identification of genes related to this process may help to reduce mortality rates through early diagnosis and the development of new anticancer therapies. Nucleophosmin 1 (NPM1) acts in ribosome biogenesis, centrosome duplication, maintenance of genomic stability, and embryonic development. Recently, NPM1 has been implicated in the tumorigenesis processes. Here, we evaluated NPM1 gene and protein expression in gastric tumors and in corresponding non-neoplastic gastric samples. Methods NPM1 protein expression was determined by Western blot in 17 pairs of gastric tumors and corresponding non-neoplastic gastric tissue. The protein immunoreactivity was observed in 12 tumor samples. mRNA expression was evaluated by reverse transcription quantitative polymerase chain reaction (RT-qPCR) in 22 pairs of gastric tumors and in matched non-neoplastic gastric tissue. Results NPM1 protein expression was significantly reduced in gastric cancer samples compared to matched non-neoplastic gastric samples (P = 0.019). The protein level of NPM1 was reduced at least 1.5-fold in 35% of tumors compared to paired non-neoplastic gastric tissue. However, NPM1 immunoreactivity was detected in neoplastic and non-neoplastic cells, including in intestinal metaplastic, gastritis and inflammatory cells. NPM1 was mainly expressed in nucleus and nucleolus subcellular compartments. The staining intensity and the percentage of immunoreactive cells varied among the studied cases. The NPM1 mRNA level was reduced at least 1.5-fold in 45.5% of samples and increased in 27.3% of samples. An inverse correlation between protein and mRNA expression was detected (r = -0.509, P = 0.037). Intestinal-type gastric cancer presented higher mRNA levels than diffuse-type (P = 0.026). However, reduced NPM1 protein expression was associated with intestinal-type gastric cancer compared to matched non-neoplastic gastric samples (P = 0.018). In addition, tumors from patients with known distant metastasis presented reduced NPM1 protein levels compared to tumors from patients without distant metastasis (P < 0.001). Conclusion Although the expression of NPM1 is heterogeneous in gastric tumors, our results suggest that NPM1 down-regulation may have a role in gastric carcinogenesis and may help in the selection of anticancer treatment strategies.
Collapse
Affiliation(s)
- Mariana Ferreira Leal
- Genetics Division, Department of Morphology and Genetic, Federal University of São Paulo, R, Botucatu, 740, São Paulo, SP CEP 04023-900, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Tsuda Y, Mori Y, Abe T, Yamashita T, Okamoto T, Ichimura T, Moriishi K, Matsuura Y. Nucleolar Protein B23 Interacts with Japanese Encephalitis Virus Core Protein and Participates in Viral Replication. Microbiol Immunol 2013; 50:225-34. [PMID: 16547420 DOI: 10.1111/j.1348-0421.2006.tb03789.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Japanese encephalitis virus (JEV) core protein is detected not only in the cytoplasm but also in the nucleoli of infected cells. We previously showed that a mutant JEV lacking the nucleolar localization of the core protein impaired viral replication in mammalian cells. In this study, we identified a nucleolar phosphoprotein B23 as a protein binding with the core protein of JEV but not with that of dengue virus. The region binding with JEV core protein was mapped to amino acid residues 38 to 77 of B23. Upon JEV infection, some fraction of B23 was translocated from the nucleoli to the cytoplasm, and cytoplasmic B23 was colocalized with the core protein of wild-type JEV but not with that of the mutant JEV. Furthermore, overexpression of dominant negatives of B23 reduced JEV replication. These results suggest that B23 plays an important role in the intracellular localization of the core protein and replication of JEV.
Collapse
Affiliation(s)
- Yoshimi Tsuda
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Oliveira AP, Simabuco FM, Tamura RE, Guerrero MC, Ribeiro PGG, Libermann TA, Zerbini LF, Ventura AM. Human respiratory syncytial virus N, P and M protein interactions in HEK-293T cells. Virus Res 2013; 177:108-12. [PMID: 23892143 DOI: 10.1016/j.virusres.2013.07.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 07/06/2013] [Accepted: 07/11/2013] [Indexed: 01/09/2023]
Abstract
Characterization of Human Respiratory Syncytial Virus (HRSV) protein interactions with host cell components is crucial to devise antiviral strategies. Viral nucleoprotein, phosphoprotein and matrix protein genes were optimized for human codon usage and cloned into expression vectors. HEK-293T cells were transfected with these vectors, viral proteins were immunoprecipitated, and co-immunoprecipitated cellular proteins were identified through mass spectrometry. Cell proteins identified with higher confidence scores were probed in the immunoprecipitation using specific antibodies. The results indicate that nucleoprotein interacts with arginine methyl-transferase, methylosome protein and Hsp70. Phosphoprotein interacts with Hsp70 and tropomysin, and matrix with tropomysin and nucleophosmin. Additionally, we performed immunoprecipitation of these cellular proteins in cells infected with HRSV, followed by detection of co-immunoprecipitated viral proteins. The results indicate that these interactions also occur in the context of viral infection, and their potential contribution for a HRSV replication model is discussed.
Collapse
Affiliation(s)
- Andressa P Oliveira
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Zidan M, Shaaban H, El Ghannam D. Prognostic Impact of Nucleophosmin 1 (NPM1) Gene Mutations in Egyptian Acute Myeloid Leukemia Patients. Turk J Haematol 2013; 30:129-36. [PMID: 24385775 PMCID: PMC3878472 DOI: 10.4274/tjh.2012.0048] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 10/02/2012] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Somatic mutations of the nucleophosmin gene (NPM1), which alter the subcellular localization of the product, are the most frequent mutations in patients with acute myeloid leukemia. The aim of the study was to assess the prevalence and prognostic impact of NPM1 gene mutations in adult AML patients. MATERIALS AND METHODS Polymerase chain reaction and single-strand conformation polymorphism (PCR-SSCP) were used to screen 55 AML patients for mutations of NPM1 gene. RESULTS NPM1 mutations were found in 12 (21.8%) of 55 patients, significantly associated with higher total leukocytie count, marrow blast percentage (p=0.03 and p=0.02, respectively), and M5 subtype (p<0.001). Patients with NPM1 mutations had significantly higher complete remission rates (p=0.003) and a trend to lower rates of mortality, relapse and refractory disease (p=0.28, p=0.45 and p=0.08, respectively). Survival analysis showed significantly longer disease-free survival (mean 18.635±1.229 versus 11.041±1.250 months, p=0.044) and overall survival (mean 19.810±1.624 versus 12.063±1.244 months, p=0.041) in patients with NPM1 mutations compared with those without. Multivariate analyses confirmed NPM1 mutation as a significant independent predictor for disease-free survival (HR=0.066, p=0.001) and overall survival (HR=0.125, p=0.002). CONCLUSION NPM1 mutation is a prognostic factor for a favorable outcome in Egyptian population. This finding is of major clinical importance since it strongly suggests that NPM1 mutations may allow one to divide the heterogeneous patient group of AML into prognostically different subgroups. CONFLICT OF INTEREST None declared.
Collapse
Affiliation(s)
- Magda Zidan
- Banha University Faculty of Medicine, Department of Clinical Pathology, Benha, Egypt
| | - Howyda Shaaban
- Banha University Faculty of Medicine, Department of Clinical Pathology, Benha, Egypt
| | - Doaa El Ghannam
- Mansoura University Faculty of Medicine, Department of Clinical Pathology, Mansoura, Egypt
| |
Collapse
|