1
|
Farias JO, Pacheco DRDCG, Magalhaes YT, Russo LC, Boell VK, Hilares DJF, Forti FL. Knockdown of dual-specificity phosphatase 3 drives differentiation and polarization of myeloid leukemia cells into macrophages with reduced proliferative and DNA repair fitness. Tissue Cell 2025; 96:102947. [PMID: 40334397 DOI: 10.1016/j.tice.2025.102947] [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: 01/15/2025] [Revised: 04/08/2025] [Accepted: 04/28/2025] [Indexed: 05/09/2025]
Abstract
Dual-specificity phosphatase 3 (DUSP3) regulates key cellular processes, including the cell cycle, proliferation, and differentiation. Recently, we demonstrated its crucial role in maintaining genomic stability by interacting with and dephosphorylating nucleophosmin (NPM), thereby modulating nuclear p53 activity under genotoxic stress. Given the frequent mutations in both p53 and NPM in acute myeloid leukemia (AML), this study aimed to investigate the impact of DUSP3 knockdown in two p53-deficient AML cell lines and explore potential correlations with NPM expression. THP-1 cells exhibited higher basal levels of DUSP3 and NPM compared to HL-60 cells, while DUSP3 knockdown reduced NPM expression in HL-60 cells. Upon phorbol 12-myristate 13-acetate (PMA)-induced differentiation into macrophage-like cells, only HL-60 cells displayed decreased levels of both DUSP3 and NPM. DUSP3 knockdown enhanced differentiation in THP-1 and HL-60 cells and promoted non-classical M2 macrophage polarization following additional PMA exposure, as indicated by increased expression of CD11b and CD206. Bioinformatics analysis revealed significant correlations between DUSP3 and NPM gene expression, AML patient survival, and the maturation stage of myeloid cells. Furthermore, DUSP3 knockdown in undifferentiated HL-60 cells impaired proliferation and compromised genomic stability under genotoxic stress induced by doxorubicin. These findings suggest that DUSP3 plays a regulatory role in the differentiation, polarization, and proliferation of myeloid cells. Through the modulation of NPM expression and activity, DUSP3 may contribute to a deeper understanding of leukemia pathophysiology and mechanisms of chemotherapy resistance.
Collapse
Affiliation(s)
- Jessica O Farias
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo, Sao Paulo, SP, Brazil
| | - Diana R D C G Pacheco
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo, Sao Paulo, SP, Brazil
| | - Yuli T Magalhaes
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo, Sao Paulo, SP, Brazil
| | - Lilian C Russo
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo, Sao Paulo, SP, Brazil
| | - Viktor K Boell
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo, Sao Paulo, SP, Brazil
| | - Donna J F Hilares
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo, Sao Paulo, SP, Brazil
| | - Fabio L Forti
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
2
|
Yan R, Tan HW, Zhao XY, Wu JY, Zhong QH, Wang XY, Cai NL, Xu YM, Lau ATY. A methionine/aspartate-rich synthetic peptide delineated from N-terminal region of nucleophosmin protein effectively protects against cadmium-induced toxicity. ENVIRONMENT INTERNATIONAL 2025; 199:109443. [PMID: 40273557 DOI: 10.1016/j.envint.2025.109443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 04/04/2025] [Accepted: 04/04/2025] [Indexed: 04/26/2025]
Abstract
Cadmium (Cd) is a widespread toxic heavy metal, and exposure to Cd is a growing environmental health concern. The molecular mechanism of Cd cytotoxicity is complicated and still not well understood, and treatment options for Cd cytotoxicity are lacking. Currently, only a limited number of Cd-targeted proteins have been identified. Here, we used Cd-immobilized metal ion affinity chromatography (Cd-IMAC) coupled with LC-MS/MS technique to detect putative Cd-binding proteins in human cells, and nucleophosmin (NPM1) was identified as the top Cd-binding protein. We found that Cd bound exclusively to the methionine/aspartate (M/D)-rich region (MEDSMDMDM) of NPM1, and NPM1 was essential for Cd-induced apoptosis. Furthermore, Cd could trigger intracellular nucleolar stress by causing nucleoplasmic translocation of NPM1 and decreasing pre-rRNA levels through binding to the M/D-rich region of NPM1. Interestingly, we discovered that a short peptide containing only the M/D-rich region of NPM1 could effectively mitigate Cd toxicity, both in vitro and in vivo. Specifically, the synthetic M/D-rich peptide demonstrated significant protection against Cd toxicity, particularly in the liver. It significantly reduced Cd concentration, suppressed the upregulation of blood ALT and AST levels, and alleviated liver inflammation in Cd-exposed BALB/c mice. This study reveals a novel mechanism of Cd cytotoxicity through NPM1-regulated nucleolar stress and apoptosis. Additionally, it identifies a short peptide with strong clinical potential to counteract Cd toxicity.
Collapse
Affiliation(s)
- Rui Yan
- The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| | - Heng Wee Tan
- The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| | - Xiao-Yun Zhao
- The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| | - Jia-Yi Wu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| | - Qiu-Hua Zhong
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| | - Xiu-Yun Wang
- The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| | - Na-Li Cai
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| | - Yan-Ming Xu
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China.
| | - Andy T Y Lau
- The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China.
| |
Collapse
|
3
|
Souza PFN, Zelaya EAE, da Silva EL, Brasil-Oliveira LL, de Oliveira FL, de Moraes MEA, Montenegro RC, Mesquita FP. PepGAT, a chitinase-derived peptide, alters the proteomic profile of colorectal cancer cells and perturbs pathways involved in cancer survival. Int J Biol Macromol 2025; 299:140204. [PMID: 39848367 DOI: 10.1016/j.ijbiomac.2025.140204] [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: 12/13/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
Colorectal cancer (CRC) affects the population worldwide, occupying the first place in terms of death and incidence. Synthetic peptides (SPs) emerged as alternative molecules due to their activity and low toxicity. Proteomic analysis of PepGAT-treated HCT-116 cells revealed a decreased abundance of proteins involved in ROS metabolism and energetic metabolisms, cell cycle, DNA repair, migration, invasion, cancer aggressiveness, and proteins involved in resistance to 5-FU. PepGAT induced earlier ROS and apoptosis in HCT-116 cells, cell cycle arrest, and inhibited HCT-116 migration. PepGAT enhances the action of 5-FU against HCT-116 cells by dropping down 6-fold the 5-FU toward HCT-116 and reduces its toxicity for non-cancerous cells. These findings strongly suggest the multiple mechanisms of action displayed by PepGAT against CRC cells and its potential to either be studied alone or in combination with 5-FU to develop new studies against CRC and might develop new drugs against it.
Collapse
Affiliation(s)
- Pedro Filho Noronha Souza
- Laboratory of Bioinformatics Applied to Health, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil; Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil; Cearense Foundation to Support Scientific and Technological Development, Brazil.
| | - Elmer Adilson Espino Zelaya
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Emerson Lucena da Silva
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Laís Lacerda Brasil-Oliveira
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Francisco Laio de Oliveira
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Maria Elisabete Amaral de Moraes
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Raquel Carvalho Montenegro
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Felipe Pantoja Mesquita
- Laboratory of Bioinformatics Applied to Health, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil; Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil.
| |
Collapse
|
4
|
Kivrak M, Nalkiran I, Sevim Nalkiran H. Exploring the Therapeutic Potential of the DOT1L Inhibitor EPZ004777 Using Bioinformatics and Molecular Docking Approaches in Acute Myeloid Leukemia. Curr Issues Mol Biol 2025; 47:173. [PMID: 40136427 PMCID: PMC11941229 DOI: 10.3390/cimb47030173] [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: 01/28/2025] [Revised: 02/19/2025] [Accepted: 03/03/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a malignancy characterized by the clonal expansion of hematopoietic stem and progenitor cells, often associated with mutations such as NPM1. DOT1L inhibitors have shown potential as new therapeutic opportunities for NPM1-mutant AML. The aim of this study was to investigate potential alternative targets of the small-molecule inhibitor EPZ004777, in addition to its primary target, DOT1L, using RNA sequencing data from the NCBI-GEO database (GSE85107). METHODS Differentially expressed genes (DEGs) were identified through bioinformatic analysis, followed by pathway enrichment analysis to uncover the relevant biological pathways. Additionally, molecular docking analysis was conducted to assess the binding affinity of EPZ004777 with the proteins CT45A3, HOXA4, SNX19, TPBG, and ZNF185, which were identified as significantly DEGs. The protein structures were obtained from AlphaFold and the Protein Data Bank. RESULTS EPZ004777 significantly altered gene expression. Oncofetal genes (CT45A3, TPBG) and genes associated with oncogenic pathways (HOXA4, ZNF185, SNX19) were downregulated, while the pro-apoptotic gene BEX3 was upregulated. Pathway enrichment analysis revealed the suppression of the Rap1 signaling pathway and cell adhesion molecules, which may reduce the invasiveness of AML cells. Additionally, upregulation of immune-related pathways suggests enhanced anti-tumor immune responses. Molecular docking analysis demonstrated that EPZ004777 has strong binding potential with SNX19, TPBG, and ZNF185 proteins. CONCLUSIONS EPZ004777 has been identified as a potent modulator of SNX19, TPBG, and ZNF185 associated with apoptosis and tumor progression in AML.
Collapse
Affiliation(s)
- Mehmet Kivrak
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Recep Tayyip Erdogan University, 53020 Rize, Türkiye
| | - Ihsan Nalkiran
- Department of Medical Biology, Faculty of Medicine, Recep Tayyip Erdogan University, 53020 Rize, Türkiye; (I.N.); (H.S.N.)
| | - Hatice Sevim Nalkiran
- Department of Medical Biology, Faculty of Medicine, Recep Tayyip Erdogan University, 53020 Rize, Türkiye; (I.N.); (H.S.N.)
| |
Collapse
|
5
|
Kendrick TS, Buic D, Fuller KA, Erber WN. Abnormalities in Chromosomes 5 and 7 in Myelodysplastic Syndrome and Acute Myeloid Leukemia. Ann Lab Med 2025; 45:133-145. [PMID: 39774131 PMCID: PMC11788707 DOI: 10.3343/alm.2024.0477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/17/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
Chromosomes 5 and 7 are large chromosomes that contain close to 1,000 genes each. Deletions of the long arms or loss of the entire chromosome (monosomy) are common defects in myeloid disorders, particularly MDS and AML. Loss of material from either chromosome 5 or 7 results in haploinsufficiency of multiple genes, with some implicated in leukemogenesis. Abnormalities of one or both occur in up to 15% of MDS and AML cases and co-segregate in half of these. Generally, these chromosomal abnormalities are harbingers of adverse risk in both myeloid disorders. A notable exception is del(5q) in 5q- syndrome, a subtype of MDS. In this review, we describe the pathogenesis and genetic consequences of deletions in chromosomes 5 and 7. Furthermore, we provide an overview of current testing methodologies used in the assessment of these chromosomal defects in hematological malignancies and describe the disease associations and prognostic implications of aberrations in chromosomes 5 and 7 in both MDS and AML.
Collapse
MESH Headings
- Humans
- Myelodysplastic Syndromes/genetics
- Myelodysplastic Syndromes/diagnosis
- Myelodysplastic Syndromes/pathology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/pathology
- Chromosomes, Human, Pair 5/genetics
- Chromosomes, Human, Pair 7/genetics
- Chromosome Deletion
- Chromosome Aberrations
Collapse
Affiliation(s)
- Tulene S. Kendrick
- Haematology Department, Royal Perth Hospital, Perth, Australia
- School of Biomedical Sciences, The University of Western Australia, Crawley, Australia
- PathWest Laboratory Medicine WA, Perth, Australia
| | - Daria Buic
- School of Biomedical Sciences, The University of Western Australia, Crawley, Australia
| | - Kathy A. Fuller
- School of Biomedical Sciences, The University of Western Australia, Crawley, Australia
| | - Wendy N. Erber
- School of Biomedical Sciences, The University of Western Australia, Crawley, Australia
- PathWest Laboratory Medicine WA, Perth, Australia
| |
Collapse
|
6
|
Ma X, Xu J, Wang Y, Fleishman JS, Bing H, Yu B, Li Y, Bo L, Zhang S, Chen ZS, Zhao L. Research progress on gene mutations and drug resistance in leukemia. Drug Resist Updat 2025; 79:101195. [PMID: 39740374 DOI: 10.1016/j.drup.2024.101195] [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: 10/03/2024] [Revised: 12/05/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025]
Abstract
Leukemia is a type of blood cancer characterized by the uncontrolled growth of abnormal cells in the bone marrow, which replace normal blood cells and disrupt normal blood cell function. Timely and personalized interventions are crucial for disease management and improving survival rates. However, many patients experience relapse following conventional chemotherapy, and increasing treatment intensity often fails to improve outcomes due to mutated gene-induced drug resistance in leukemia cells. This article analyzes the association of gene mutations and drug resistance in leukemia. It explores genetic abnormalities in leukemia, highlighting recently identified mutations affecting signaling pathways, cell apoptosis, epigenetic regulation, histone modification, and splicing mechanisms. Additionally, the article discusses therapeutic strategies such as molecular targeting of gene mutations, alternative pathway targeting, and immunotherapy in leukemia. These approaches aim to combat specific drug-resistant mutations, providing potential avenues to mitigate leukemia relapse. Future research with these strategies holds promise for advancing leukemia treatment and addressing the challenges of drug-resistant mutations to improve patient outcomes.
Collapse
Affiliation(s)
- Xiangyu Ma
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China
| | - Jiamin Xu
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
| | - Yanan Wang
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY 11439, USA
| | - Hao Bing
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China
| | - Boran Yu
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China
| | - Yanming Li
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China
| | - Letao Bo
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY 11439, USA
| | - Shaolong Zhang
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY 11439, USA.
| | - Libo Zhao
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China; Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
7
|
Castaño-Díez S, Álamo JR, López-Guerra M, Gómez-Hernando M, Zugasti I, Jiménez-Vicente C, Guijarro F, López-Oreja I, Esteban D, Charry P, Torrecillas V, Mont-de Torres L, Cortés-Bullich A, Bataller Á, Guardia A, Munárriz D, Carcelero E, Riu G, Triguero A, Tovar N, Vela D, Beà S, Costa D, Colomer D, Rozman M, Esteve J, Díaz-Beyá M. Chronic myelomonocytic leukemia with NPM1 mutation or acute myeloid leukemia? Oncologist 2025; 30:oyae246. [PMID: 39349391 PMCID: PMC11884738 DOI: 10.1093/oncolo/oyae246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 04/20/2024] [Indexed: 10/02/2024] Open
Abstract
The 2022 WHO revision and the ICC classification have recently modified the diagnostic criteria for chronic myelomonocytic leukemia (CMML) and acute myeloid leukemia. However, there is no consensus on whether CMML with NPM1 mutation (NPM1mut) should be diagnosed as AML. Nowadays, it is a subject of discussion because of its diagnostic and therapeutic implications. Therefore, we describe a case of a patient diagnosed with CMML NPM1mut and briefly review the literature to highlight the uncertainty about how to classify a CMML with NPM1 mutation. We emphasize the importance of a comprehensive molecular study, which is crucial to optimize the individualized treatment of patients, enabling them to access targeted therapies.
Collapse
MESH Headings
- Humans
- Nucleophosmin
- Leukemia, Myelomonocytic, Chronic/genetics
- Leukemia, Myelomonocytic, Chronic/diagnosis
- Leukemia, Myelomonocytic, Chronic/pathology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/pathology
- Mutation
- Nuclear Proteins/genetics
- Male
- Aged
- Female
Collapse
Affiliation(s)
- Sandra Castaño-Díez
- Hematology Department, Hospital Clínic Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Universitat Barcelona, Barcelona, Spain
| | - José Ramón Álamo
- Hematopathology Section, Pathology Department, Hospital Clínic Barcelona, Barcelona, Spain
| | - Mònica López-Guerra
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Hematopathology Section, Pathology Department, Hospital Clínic Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Marta Gómez-Hernando
- Hematopathology Section, Pathology Department, Hospital Clínic Barcelona, Barcelona, Spain
| | - Inés Zugasti
- Hematology Department, Hospital Clínic Barcelona, Barcelona, Spain
| | | | - Francesca Guijarro
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Universitat Barcelona, Barcelona, Spain
- Hematopathology Section, Pathology Department, Hospital Clínic Barcelona, Barcelona, Spain
| | - Irene López-Oreja
- Hematopathology Section, Pathology Department, Hospital Clínic Barcelona, Barcelona, Spain
| | - Daniel Esteban
- Hematology Department, Hospital Clínic Barcelona, Barcelona, Spain
| | - Paola Charry
- Hematology Department, Hospital Clínic Barcelona, Barcelona, Spain
| | | | | | | | - Álex Bataller
- Hematology Department, Hospital Clínic Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Universitat Barcelona, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Badalona, Spain
| | - Ares Guardia
- Hematology Department, Hospital Clínic Barcelona, Barcelona, Spain
| | - Daniel Munárriz
- Hematology Department, Hospital Clínic Barcelona, Barcelona, Spain
| | | | - Gisela Riu
- Pharmacy Service, Hospital Clínic Barcelona, Barcelona, Spain
| | - Ana Triguero
- Hematology Department, Hospital Clínic Barcelona, Barcelona, Spain
| | - Natalia Tovar
- Hematology Department, Hospital Clínic Barcelona, Barcelona, Spain
| | - Dolors Vela
- Hematology Department, Hospital General de Granollers, Granollers, Spain
| | - Silvia Beà
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Hematopathology Section, Pathology Department, Hospital Clínic Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Dolors Costa
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Hematopathology Section, Pathology Department, Hospital Clínic Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Dolors Colomer
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Hematopathology Section, Pathology Department, Hospital Clínic Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Maria Rozman
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Hematopathology Section, Pathology Department, Hospital Clínic Barcelona, Barcelona, Spain
| | - Jordi Esteve
- Hematology Department, Hospital Clínic Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Universitat Barcelona, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Badalona, Spain
| | - Marina Díaz-Beyá
- Hematology Department, Hospital Clínic Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Badalona, Spain
| |
Collapse
|
8
|
Chen T, Chen H, Xia M, Liao Y, Li H, Dong X, Lin Y, Zhou W. In-depth inference of transcriptional regulatory networks reveals NPM1 as a therapeutic ribosomal regulator in MYC-amplified medulloblastoma. NPJ Precis Oncol 2025; 9:10. [PMID: 39794402 PMCID: PMC11723958 DOI: 10.1038/s41698-024-00792-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/19/2024] [Indexed: 01/13/2025] Open
Abstract
Medulloblastoma (MB) is an aggressive pediatric brain tumor with distinct molecular heterogeneity. Identifying subtype-specific signatures within Group 3 and Group 4 remains challenging due to shared cytogenetic alterations and limitations of conventional differential gene expression analysis. To uncover the underlying molecular signatures and hidden regulators, we used the Cavalli transcriptomic profile of 470 Group 3 and Group 4 MB patients to reconstruct subtype-specific regulatory networks. A strong upregulation of the ribosomal pathway was linked to MYC amplification in Group 3, with Nucleophosmin 1 (NPM1) emerging as a key regulator. NPM1 upregulation defined a subset of Group3 and Group4 patients with poor prognosis. Inhibition of NPM1 led to apoptosis, reduced c-Myc stability, and impaired translation in MYC-amplified Group 3 MB cells. Together, our findings highlight NPM1 as a promising therapeutic target and provide new insights into the regulatory mechanisms in MB.
Collapse
Affiliation(s)
- Tong Chen
- Key Laboratory of Neonatal Disease, Ministry of Health, Children's Hospital of Fudan University, Shanghai, China
| | - Huiyao Chen
- Center for Molecular Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Mingyang Xia
- Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Fudan University, Shanghai, China
| | - Yunfei Liao
- Center for Molecular Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Hao Li
- Department of Neurosurgery, Children's Hospital of Fudan University, Shanghai, China
| | - Xinran Dong
- Center for Molecular Medicine, Children's Hospital of Fudan University, Shanghai, China.
| | - Yifeng Lin
- Key Laboratory of Neonatal Disease, Ministry of Health, Children's Hospital of Fudan University, Shanghai, China.
| | - Wenhao Zhou
- Division of Neonatology and Center for Newborn Care, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
9
|
Ali A, Paracha S, Pincus D. Preserve or destroy: Orphan protein proteostasis and the heat shock response. J Cell Biol 2024; 223:e202407123. [PMID: 39545954 PMCID: PMC11572482 DOI: 10.1083/jcb.202407123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024] Open
Abstract
Most eukaryotic genes encode polypeptides that are either obligate members of hetero-stoichiometric complexes or clients of organelle-targeting pathways. Proteins in these classes can be released from the ribosome as "orphans"-newly synthesized proteins not associated with their stoichiometric binding partner(s) and/or not targeted to their destination organelle. Here we integrate recent findings suggesting that although cells selectively degrade orphan proteins under homeostatic conditions, they can preserve them in chaperone-regulated biomolecular condensates during stress. These orphan protein condensates activate the heat shock response (HSR) and represent subcellular sites where the chaperones induced by the HSR execute their functions. Reversible condensation of orphan proteins may broadly safeguard labile precursors during stress.
Collapse
Affiliation(s)
- Asif Ali
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| | - Sarah Paracha
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| | - David Pincus
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
- Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA
- Center for Physics of Evolution, University of Chicago, Chicago, IL, USA
| |
Collapse
|
10
|
Ishikawa Y, Ushijima Y, Kiyoi H. Recent advances in AML with mutated NPM1. Int J Hematol 2024; 120:556-565. [PMID: 39174699 DOI: 10.1007/s12185-024-03835-8] [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: 06/28/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/24/2024]
Abstract
Nucleophosmin 1 (NPM1) mutation is one of the most prevalent genetic mutations in adult acute myeloid leukemia (AML) and is particularly predominant in AML with a normal karyotype. NPM1 is a chaperone protein that plays various roles in several cellular processes. Wild-type NPM1 is normally localized to the nucleus, whereas mutant NPM1 proteins exhibit altered cytoplasmic localization. Clinically, AML with mutated NPM1 without FLT3-ITD is associated with a higher complete remission rate and improved overall survival. AML with mutated NPM1 is categorized as a distinct genetic entity in the World Health Organization classification of hematopoietic malignancies due to its unique clinical and biological features. However, the precise roles of NPM1 in normal hematopoiesis and in AML development remain unclear. Recent studies have revealed various clinical applications of NPM1 mutations in AML treatment, particularly in measurable residual disease analyses that target mutant NPM1 transcripts and in potential therapeutic applications of menin inhibitors and XPO-1 inhibitors for AML with mutated NPM1. Thus, NPM1 mutation is highly significant in AML classification, prognosis, response assessment, and molecular targeted therapies. Here, we review recent progress in clinical and biological aspects of AML with mutated NPM1 including molecular targeted therapy.
Collapse
Affiliation(s)
- Yuichi Ishikawa
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan.
| | - Yoko Ushijima
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| |
Collapse
|
11
|
Smith E, Atenafu EG, Bankar A, Chan S, Davidson M, Gupta V, Minden MD, Richard-Carpentier G, Schimmer A, Schuh AC, Sibai H, Yee K, Maze D. Evolution from an antecedent chronic myeloid malignancy does not impact survival outcomes in NPM1-mutated AML. Eur J Haematol 2024; 113:716-726. [PMID: 39113600 DOI: 10.1111/ejh.14283] [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: 04/10/2024] [Revised: 07/17/2024] [Accepted: 07/20/2024] [Indexed: 10/03/2024]
Abstract
Nucleophosmin-1 (NPM1)-mutated AML is a molecularly defined subtype typically associated with favorable treatment response and prognosis; however, its prognostic significance in AML evolving from an antecedent chronic myeloid malignancy is unknown. This study's primary objective was to determine the impact of mutated NPM1 on the prognosis of AML evolving from an antecedent chronic myeloid malignancy. We conducted a retrospective chart review including patients with NPM1-mutated de novo and sAML. sAML was defined as those with a preceding chronic-phase myeloid malignancy before diagnosis of AML. Of 575 NPM1-mutated patients eligible for inclusion in our study, 51 (8.9%) patients were considered to have sAML. The median time from diagnosis of NPM1-mutated chronic myeloid malignancy to sAML evolution was 3.6 months (0.5-79.3 months). No significant differences in leukemia-free (2-year LKFS 52.0% vs. 51.2%, p = .9922) or overall survival (2-year OS 56.3% vs. 49.4%, p = .4246) were observed between patients with NPM1-mutated de novo versus sAML. Our study suggests that evolution from a preceding myeloid malignancy is not a significant predictor of poor prognosis in the setting of an NPM1 mutation. Our study demonstrated a short time to progression to sAML in most patients, which further supports the consideration of NPM1 as an AML-defining mutation.
Collapse
MESH Headings
- Humans
- Nucleophosmin
- Mutation
- Nuclear Proteins/genetics
- Male
- Female
- Middle Aged
- Aged
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/diagnosis
- Prognosis
- Retrospective Studies
- Adult
- Aged, 80 and over
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis
- Clonal Evolution/genetics
Collapse
Affiliation(s)
- Elliot Smith
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Eshetu G Atenafu
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Aniket Bankar
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Steven Chan
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Marta Davidson
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Vikas Gupta
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Mark D Minden
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Guillaume Richard-Carpentier
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Aaron Schimmer
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Andre C Schuh
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Hassan Sibai
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Karen Yee
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Dawn Maze
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
12
|
Gener-Ricos G, Bataller A, Rodriguez-Sevilla JJ, Chien KS, Quesada AE, Almanza-Huante E, Hammond D, Sasaki K, DiNardo C, Kadia T, Daver N, Borthakur G, Issa GC, Short NJ, Kanagal-Shamanna R, Kantarjian HM, Garcia-Manero G, Montalban-Bravo G. NPM1-mutated myeloid neoplasms are a unique entity not defined by bone marrow blast percentage. Cancer 2024; 130:3452-3462. [PMID: 38896064 DOI: 10.1002/cncr.35433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/02/2024] [Accepted: 05/14/2024] [Indexed: 06/21/2024]
Abstract
INTRODUCTION NPM1-mutated (NPM1mut) myeloid neoplasms (MNs) with <20% bone marrow (BM) blasts (NPM1mut MNs<20) are uncommon, and their classification remains inconsistent. METHODS The clinicopathologic features of 54 patients with NPM1mut MNs <20 were evaluated and compared with wild-type NPM1 MNs <20 and NPM1mut MNs≥20, respectively. RESULTS NPM1mut MNs had similar features regardless of blast percentage, except for higher IDH2 (29% vs 7%, p = .023) and FLT3 (70% vs 11%, p < .001) frequency in patients with ≥20% BM blasts. Thirty-three (61%) patients with NPM1mut MNs <20 received low-intensity chemotherapy (LIC) and 12 (22%) received intensive chemotherapy (IC). Higher complete remission rates (75% vs 27%, p = .006) and median overall survival (mOS) (not reached vs 30.4 months, p = .06) were observed with IC compared to LIC. Young patients (age <60 years) did not reach mOS either when treated with LIC or IC. Stem cell transplant was associated with increased survival only in patients treated with LIC (HR, 0.24; p = .025). No differences in mOS were observed by BM blast strata (32.2 months, not reached and 46.9 months for <10%, 10%-19%, and ≥20% blasts, p = .700) regardless of treatment modality (LIC: p = .900; IC: p = .360). Twenty-three patients (43%) with NPM1mut MNs <20 had marrow blast progression to ≥20%. CONCLUSIONS Overall, NPM1mut MNs define a unique entity independent of BM blast percentage.
Collapse
Affiliation(s)
- Georgina Gener-Ricos
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Alex Bataller
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Kelly S Chien
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Andres E Quesada
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Emmanuel Almanza-Huante
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Danielle Hammond
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Koji Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Courtney DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tapan Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ghayas C Issa
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nicholas J Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Guillermo Garcia-Manero
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | |
Collapse
|
13
|
Wang X, Chai Y, Quan Y, Wang J, Song J, Zhou W, Xu X, Xu H, Wang B, Cao X. NPM1 inhibits tumoral antigen presentation to promote immune evasion and tumor progression. J Hematol Oncol 2024; 17:97. [PMID: 39402629 PMCID: PMC11479574 DOI: 10.1186/s13045-024-01618-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Tumor cells develop multiple mechanisms to facilitate their immune evasion. Identifying tumor-intrinsic factors that support immune evasion may provide new strategies for cancer immunotherapy. We aimed to explore the function and the mechanism of the tumor-intrinsic factor NPM1, a multifunctional nucleolar phosphoprotein, in cancer immune evasion and progression. METHODS The roles of NPM1 in tumor progression and tumor microenvironment (TME) reprogramming were examined by subcutaneous inoculation of Npm1-deficient tumor cells into syngeneic mice, and then explored by CyTOF, flow cytometry, immunohistochemistry staining, and RNA-seq. The in-vitro T-cell killing of OVA-presenting tumor cells by OT-1 transgenic T cells was observed. The interaction of NPM1 and IRF1 was verified by Co-IP. The regulation of NPM1 in IRF1 DNA binding to Nlrc5, Ciita promoter was determined by dual-luciferase reporter assay and ChIP-qPCR. RESULTS High levels of NPM1 expression predict low survival rates in various human tumors. Loss of NPM1 inhibited tumor progression and enhanced the survival of tumor-bearing mice. Npm1-deficient tumors showed increased CD8+ T cell infiltration and activation alongside the reduced presence of immunosuppressive cells. Npm1 deficiency increased MHC-I and MHC-II molecules and specific T-cell killing. Mechanistically, NPM1 associates with the transcription factor IRF1 and then sequesters IRF1 from binding to the Nlrc5 and Ciita promoters to suppress IRF1-mediated expression of MHC-I and MHC-II molecules in tumor cells. CONCLUSIONS Tumor-intrinsic NPM1 promotes tumor immune evasion via suppressing IRF1-mediated antigen presentation to impair tumor immunogenicity and reprogram the immunosuppressive TME. Our study identifies NPM1 as a potential target for improving cancer immunotherapy.
Collapse
Affiliation(s)
- Xin Wang
- Department of Immunology, Center for Immunotherapy, Institute of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Yangyang Chai
- Department of Immunology, Center for Immunotherapy, Institute of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Yuan Quan
- Department of Immunology, Center for Immunotherapy, Institute of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Jiaming Wang
- Department of Immunology, Center for Immunotherapy, Institute of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Jiaying Song
- Department of Immunology, Center for Immunotherapy, Institute of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Wenkai Zhou
- Department of Immunology, Center for Immunotherapy, Institute of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Xiaoqing Xu
- Department of Immunology, Center for Immunotherapy, Institute of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Henan Xu
- Frontier Research Center for Cell Response, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Bingjing Wang
- Department of Immunology, Center for Immunotherapy, Institute of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Xuetao Cao
- Department of Immunology, Center for Immunotherapy, Institute of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China.
- Frontier Research Center for Cell Response, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
14
|
Greiner J, Mohamed E, Fletcher DM, Schuler PJ, Schrezenmeier H, Götz M, Guinn BA. Immunotherapeutic Potential of Mutated NPM1 for the Treatment of Acute Myeloid Leukemia. Cancers (Basel) 2024; 16:3443. [PMID: 39456538 PMCID: PMC11505958 DOI: 10.3390/cancers16203443] [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: 06/19/2024] [Revised: 09/15/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024] Open
Abstract
Acute myeloid leukemia (AML) is a malignant disease of the blood and bone marrow that is characterized by uncontrolled clonal proliferation of abnormal myeloid progenitor cells. Nucleophosmin 1 (NPM1) gene mutations are the most common genetic abnormality in AML, detectable in blast cells from about one-third of adults with AML. AML NPM1mut is recognized as a separate entity in the World Health Organization classification of AML. Clinical and survival data suggest that patients with this form of AML often have a more favorable prognosis, which may be due to the immunogenicity created by the mutations in the NPM1 protein. Consequently, AML with NPM1mut can be considered an immunogenic subtype of AML. However, the underlying mechanisms of this immunogenicity and associated favorable survival outcomes need to be further investigated. Immune checkpoint molecules, such as the programmed cell death-1 (PD-1) protein and its ligand, PD-L1, play important roles in leukemogenesis through their maintenance of an immunosuppressive tumor microenvironment. Preclinical trials have shown that the use of PD-1/PD-L1 checkpoint inhibitors in solid tumors and lymphoma work best in novel therapy combinations. Patients with AML NPM1mut may be better suited to immunogenic strategies that are based on the inhibition of the PD-1 immune checkpoint pathway than patients without this mutation, suggesting the genetic landscape of patients may also inform best practice for the use of PD-1 inhibitors.
Collapse
Affiliation(s)
- Jochen Greiner
- Department of Internal Medicine III, University Hospital Ulm, 89081 Ulm, Germany;
- Department of Internal Medicine, Diakonie Hospital Stuttgart, 70176 Stuttgart, Germany
| | - Eithar Mohamed
- Centre for Biomedicine, Hull York Medical School, University of Hull, Hull HU6 7RX, UK; (E.M.); (D.M.F.)
| | - Daniel M. Fletcher
- Centre for Biomedicine, Hull York Medical School, University of Hull, Hull HU6 7RX, UK; (E.M.); (D.M.F.)
| | - Patrick J. Schuler
- Department of Otorhinolaryngology, University Hospital Ulm, 89075 Ulm, Germany;
- Department of Oto-Rhino-Laryngology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Hubert Schrezenmeier
- Institute of Transfusion Medicine, University of Ulm, 89073 Ulm, Germany;
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, 89081 Ulm, Germany
| | - Marlies Götz
- Department of Internal Medicine III, University Hospital Ulm, 89081 Ulm, Germany;
- Department of Internal Medicine, Diakonie Hospital Stuttgart, 70176 Stuttgart, Germany
| | - Barbara-ann Guinn
- Centre for Biomedicine, Hull York Medical School, University of Hull, Hull HU6 7RX, UK; (E.M.); (D.M.F.)
| |
Collapse
|
15
|
Bangash AA, Alvi SS, Bangash MA, Ahsan H, Khan S, Shareef R, Villanueva G, Bansal D, Ahmad M, Kim DJ, Chauhan SC, Hafeez BB. Honey Targets Ribosome Biogenesis Components to Suppress the Growth of Human Pancreatic Cancer Cells. Cancers (Basel) 2024; 16:3431. [PMID: 39410048 PMCID: PMC11475701 DOI: 10.3390/cancers16193431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 10/20/2024] Open
Abstract
Pancreatic cancer (PanCa) is one of the deadliest cancers, with limited therapeutic response. Various molecular oncogenic events, including dysregulation of ribosome biogenesis, are linked to the induction, progression, and metastasis of PanCa. Thus, the discovery of new therapies suppressing these oncogenic events and ribosome biogenesis could be a novel therapeutic approach for the prevention and treatment of PanCa. The current study was designed to investigate the anti-cancer effect of honey against PanCa. Our results indicated that honey markedly inhibited the growth and invasive characteristics of pancreatic cancer cells by suppressing the mRNA expression and protein levels of key components of ribosome biogenesis, including RNA Pol-I subunits (RPA194 and RPA135) along with its transcriptional regulators, i.e., UBTF and c-Myc. Honey also induced nucleolar stress in PanCa cells by reducing the expression of various nucleolar proteins (NCL, FBL, and NPM). Honey-mediated regulation on ribosome biogenesis components and nucleolar organization-associated proteins significantly arrested the cell cycle in the G2M phase and induced apoptosis in PanCa cells. These results, for the first time, demonstrated that honey, being a natural remedy, has the potential to induce apoptosis and inhibit the growth and metastatic phenotypes of PanCa by targeting ribosome biogenesis.
Collapse
Affiliation(s)
- Aun Ali Bangash
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Sahir Sultan Alvi
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Muhammad Ali Bangash
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Haider Ahsan
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Shiza Khan
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Rida Shareef
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Georgina Villanueva
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Divyam Bansal
- Department of Kinesiology, Rice University, Houston, TX 77251, USA;
| | - Mudassier Ahmad
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Dae Joon Kim
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Subhash C. Chauhan
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Bilal Bin Hafeez
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| |
Collapse
|
16
|
Genoveso MJ, Okuwaki M, Kato K, Nagata K, Kawaguchi A. Nuclear reorganization by NPM1-mediated phase separation triggered by adenovirus core protein VII. Microbiol Spectr 2024; 12:e0041624. [PMID: 39162498 PMCID: PMC11448090 DOI: 10.1128/spectrum.00416-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/18/2024] [Indexed: 08/21/2024] Open
Abstract
Recent evidence has revealed that the reorganization of nuclear domains is largely mediated by liquid-liquid phase separation (LLPS). During viral infection, numerous nuclear domains undergo significant changes through LLPS for and against the replication of the virus. However, the regulatory mechanism of LLPS in response to viral infection and its detailed functions in viral replication remain unclear. In this study, we found that the activity of the nucleolar protein NPM1, a remodeling factor for the chromatin-like structure of adenovirus DNA, to induce LLPS is required for deposition of adenovirus core protein VII in a subnuclear domain, the virus-induced post-replication (ViPR) body, in the late phases of infection. The interaction between NPM1 and protein VII was responsible for initiating LLPS. The inhibition of LLPS by 1,6-hexanediol treatment resulted in the dispersion of protein VII from the ViPR bodies. These findings suggest that protein VII accumulates in the ViPR bodies in concert with the LLPS formation of NPM1 triggered by protein VII. After photobleaching of EGFP-NPM1 in the ViPR bodies, EGFP-NPM1 showed a relatively fast recovery half-time, indicating the fluid-like properties of NPM1 in this compartment. Importantly, NPM1 depletion decreased the genome packaging in the viral capsids, possibly owing to the formation of a defective adenovirus core. This study highlights the dynamic interplay between viral pathogens and the host nucleus for the reorganization of membrane-less compartments that facilitate their replication. IMPORTANCE In this study, we explored how adenoviruses utilize a process known as liquid-liquid phase separation (LLPS) to enhance their replication. We focused on a cellular chromatin remodeling protein, NPM1, which plays a crucial role in nucleolar formation through LLPS. NPM1 facilitates LLPS by interacting with adenovirus protein VII, effectively accumulating protein VII into membrane-less compartments called virus-induced post-replication bodies. NPM1 functions as a molecular chaperone of protein VII to assemble viral chromatin by transferring protein VII to viral DNA. Remarkably, when NPM1 was depleted, this process was disrupted, decreasing viral genome packaging. These findings shed light on a critical aspect of virus-host interactions, illustrating how adenovirus utilizes NPM1-mediated LLPS activity. Our findings provide valuable insights into the dynamic interplay between viruses and the host nucleus.
Collapse
Affiliation(s)
- Michelle Jane Genoveso
- Department of Infection Biology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Mitsuru Okuwaki
- Laboratory of Biochemistry, School of Pharmacy, Kitasato University, Tokyo, Japan
| | - Kohsuke Kato
- Department of Infection Biology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kyosuke Nagata
- Department of Infection Biology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Atsushi Kawaguchi
- Department of Infection Biology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Transborder Medical Research Center, University of Tsukuba, Tsukuba, Japan
- Microbiology Research Center for Sustainability, University of Tsukuba, Tsukuba, Japan
- Center for Quantum and Information Life Sciences, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
17
|
Tao X, Wei H, Mao S, Wang J, Xue C, Yu W, Shi Y, Liu Y, Sun B. SEC24C suppresses the propagation and chemoresistance of hepatocellular carcinoma by promoting unfolded protein response-related apoptosis. Biosci Trends 2024; 18:343-355. [PMID: 39085101 DOI: 10.5582/bst.2024.01149] [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] [Indexed: 08/02/2024]
Abstract
Cells routinely utilize the unfolded protein response (UPR) to alleviate endoplasmic reticulum (ER)-stress or trigger about apoptotic death under extreme ER-stress conditions. Tumor cells are subjected to persistent ER-stress due to their crowded microenvironment, but can maintain hyperactive proliferation under most stressful conditions. Therefore, understanding strategies employed by cancer cells to escape from UPR-related apoptosis has important medical implications. SEC24 homolog C (SEC24C) was found decreased in later colorectal cancer (CRC) stages, but its exact role in response to ER-stress and activation of UPR in hepatocellular carcinoma (HCC) remains to be elucidated. Here, we have identified the downregulation of SEC24C in human HCC sample and its suppressive role in regulating HCC proliferation and chemoresistance. Mechanistically, SEC24C was found to interact with eukaryotic translation initiation factor 2 alpha kinase 3 (EIF2AK3 or PERK) and activate the downstream UPR-related apoptosis. During this process, SEC24C was observed to be anchored in nucleus under normal condition but responded immediately to ER-stress and could subsequently translocate to the ER. Furthermore, overexpression of SEC24C significantly augmented the efficacy of bortezomib in HCC treatment. In conclusion, our findings revealed a novel role of SEC24C in regulating HCC proliferation and chemoresistance by modulating UPR activation.
Collapse
Affiliation(s)
- Xuewen Tao
- School of Medicine, Southeast University, Nanjing, Jiangsu, China
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Haowei Wei
- The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Shuai Mao
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Lianyungang oriental hospital, Lianyungang, Jiangsu, China
| | - Jincheng Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Graduate School of Medical Science and Engineering, Hokkaido University, Sapporo, Japan
| | - Cailin Xue
- The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Weiwei Yu
- The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yuze Shi
- The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yang Liu
- School of Medicine, Southeast University, Nanjing, Jiangsu, China
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Beicheng Sun
- School of Medicine, Southeast University, Nanjing, Jiangsu, China
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
18
|
Wang T, Cui S, Lyu C, Wang Z, Li Z, Han C, Liu W, Wang Y, Xu R. Molecular precision medicine: Multi-omics-based stratification model for acute myeloid leukemia. Heliyon 2024; 10:e36155. [PMID: 39263156 PMCID: PMC11388765 DOI: 10.1016/j.heliyon.2024.e36155] [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: 02/25/2024] [Revised: 08/01/2024] [Accepted: 08/11/2024] [Indexed: 09/13/2024] Open
Abstract
Acute myeloid leukemia (AML), as the most common malignancy of the hematopoietic system, poses challenges in treatment efficacy, relapse, and drug resistance. In this study, we have utilized 151 RNA sequencing datasets, 194 DNA methylation datasets, and 200 somatic mutation datasets from the AML cohort in the TCGA database to develop a multi-omics stratification model. This model enables comparison of prognosis, clinical features, gene mutations, immune microenvironment and drug sensitivity across subgroups. External validation datasets have been sourced from the GEO database, which includes 562 mRNA datasets and 136 miRNA datasets from 984 adult AML patients. Through multi-omics-based stratification model, we classified 126 AML patients into 4 clusters (CS). CS4 had the best prognosis, with the youngest age, highest M3 subtype proportion, fewest copy number alterations, and common mutations in WT1, FLT3, and KIT genes. It showed sensitivity to HDAC inhibitors and BCL-2 inhibitors. Both the M3 subtype and CS4 were identified as independent protective factors for survival. Conversely, CS3 had the worst prognosis due to older age, high copy number alterations, and frequent mutations in RUNX1, DNMT3A, and TP53 genes. Additionally, it showed higher proportions of cytotoxic cells and Tregs, suggesting potential sensitivity to mTOR inhibitors. CS1 had a better prognosis than CS2, with more copy number alterations, while CS2 had higher monocyte proportions. CS1 showed good sensitivity to cytarabine, while CS2 was sensitive to RXR agonists. Both CS1 and CS2, which predominantly featured mutations in FLT3, NPM1, and DNMT3A genes, benefited from FLT3 inhibitors. Using the Kappa test, our stratification model underwent robust validation in the miRNA and mRNA external validation datasets. With advancements in sequencing technology and machine learning algorithms, AML is poised to transition towards multi-omics precision medicine in the future. We aspire for our study to offer new perspectives on multi-drug combination clinical trials and multi-targeted precision medicine for AML.
Collapse
Affiliation(s)
- Teng Wang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Siyuan Cui
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Hematology, Health Commission of Shandong Province, Shandong, 250014, China
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Shandong, 250014, China
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong, 250014, China
| | - Chunyi Lyu
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhenzhen Wang
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Hematology, Health Commission of Shandong Province, Shandong, 250014, China
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Shandong, 250014, China
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong, 250014, China
| | - Zonghong Li
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chen Han
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Weilin Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Wang
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Hematology, Health Commission of Shandong Province, Shandong, 250014, China
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Shandong, 250014, China
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong, 250014, China
| | - Ruirong Xu
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Hematology, Health Commission of Shandong Province, Shandong, 250014, China
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Shandong, 250014, China
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong, 250014, China
| |
Collapse
|
19
|
Wang P, Wang M, Liu L, Li H, Liu H, Ren J, Liu T, Cong M, Zhu Z, Zhao X, Sun L, Jia J. Targeting NPM1 inhibits proliferation and promotes apoptosis of hepatic progenitor cells via suppression of mTOR signalling pathway. Stem Cell Res Ther 2024; 15:292. [PMID: 39256792 PMCID: PMC11389206 DOI: 10.1186/s13287-024-03898-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 08/26/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Hepatic progenitor cells serve not only as the origin of combined hepatocellular cholangiocarcinoma (cHCC-CCA) but are also responsible for malignancy recurrence after surgical resection. Nucleophosmin 1 (NPM1) has been implicated in cancer metastasis and poor prognosis. This study aimed to determine the expression of NPM1 by hepatic progenitor cells in cHCC-CCA and the effects of targeting NPM1 on hepatic progenitor cells and BEL-7402 cells with characteristics of both progenitor cells and cHCC-CCA. METHODS First, NPM1 was detected by RT‒PCR, western blotting, and double-immunofluorescence staining in cHCC-CCA tissues. NPM1 expression was subsequently analysed in rat hepatic progenitor cells cultured in vitro and in interleukin 6 (IL6)-treated cells. The effects and mechanism of NPM1 on hepatic progenitor cells were determined by knocking down NPM1 and performing RNA sequencing analysis. Finally, NSC348884, a small-molecule inhibitor that disrupts NPM1 dimer formation, was used to confirm the function of NPM1 in BEL-7402 cells. RESULTS Both human hepatic progenitor cells in cHCC-CCA tissues and rat in vitro cultured hepatic progenitor cells highly expressed NPM1. IL6, a cytokine involved in the malignant transformation of hepatic progenitor cells, dose-dependently increased NPM1 and PCNA expression. Knocking down NPM1 reduced IL6R transcription (P < 0.0001) and inhibited the proliferation (P = 0.0065) of hepatic progenitor cells by suppressing the mTOR signalling pathway and activating the apoptosis pathway. Furthermore, knocking down NPM1 in hepatic progenitor cells resulted in more apoptotic cells (7.33 ± 0.09% vs. 3.76 ± 0.13%, P < 0.0001) but fewer apoptotic cells in the presence of NSC348884 (47.57 ± 0.49% vs. 63.40 ± 0.05%, P = 0.0008) than in the control cells, suggesting that low-NPM1-expressing cells are more resistant to NSC348884. In addition, NSC348884 induced the apoptosis of BEL-7402 cells with an IC50 of 2.77 μmol/L via the downregulation of the IL-6R and mTOR signalling pathways and inhibited the growth of BEL-7402 cells in a subcutaneous xenograft tumour model (P = 0.0457). CONCLUSIONS Targeting NPM1 inhibits proliferation and induces apoptosis in hepatic progenitor cells and BEL-7402 cells, thus serving as a potential therapy for cHCC-CCA.
Collapse
Affiliation(s)
- Ping Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
- Beijing Key Laboratory On Translational Medicine On Liver Cirrhosis, Beijing, 100050, China.
- National Clinical Research Center for Digestive Disease, Beijing, 100069, China.
| | - Min Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- Beijing Key Laboratory On Translational Medicine On Liver Cirrhosis, Beijing, 100050, China
- National Clinical Research Center for Digestive Disease, Beijing, 100069, China
| | - Lin Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- Beijing Key Laboratory On Translational Medicine On Liver Cirrhosis, Beijing, 100050, China
- National Clinical Research Center for Digestive Disease, Beijing, 100069, China
| | - Hongyi Li
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- Beijing Key Laboratory On Translational Medicine On Liver Cirrhosis, Beijing, 100050, China
- National Clinical Research Center for Digestive Disease, Beijing, 100069, China
| | - Helin Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- Beijing Key Laboratory On Translational Medicine On Liver Cirrhosis, Beijing, 100050, China
- National Clinical Research Center for Digestive Disease, Beijing, 100069, China
| | - Jiangbo Ren
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- Beijing Key Laboratory On Translational Medicine On Liver Cirrhosis, Beijing, 100050, China
- National Clinical Research Center for Digestive Disease, Beijing, 100069, China
| | - Tianhui Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- Beijing Key Laboratory On Translational Medicine On Liver Cirrhosis, Beijing, 100050, China
- National Clinical Research Center for Digestive Disease, Beijing, 100069, China
| | - Min Cong
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- Beijing Key Laboratory On Translational Medicine On Liver Cirrhosis, Beijing, 100050, China
- National Clinical Research Center for Digestive Disease, Beijing, 100069, China
| | - Zhijun Zhu
- National Clinical Research Center for Digestive Disease, Beijing, 100069, China
- Liver Transplantation Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- Clinical Center for Paediatric Liver Transplantation, Capital Medical University, Beijing, 100069, China
| | - Xinyan Zhao
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- Beijing Key Laboratory On Translational Medicine On Liver Cirrhosis, Beijing, 100050, China
- National Clinical Research Center for Digestive Disease, Beijing, 100069, China
| | - Liying Sun
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
- Beijing Key Laboratory On Translational Medicine On Liver Cirrhosis, Beijing, 100050, China.
- National Clinical Research Center for Digestive Disease, Beijing, 100069, China.
| | - Jidong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
- Beijing Key Laboratory On Translational Medicine On Liver Cirrhosis, Beijing, 100050, China.
- National Clinical Research Center for Digestive Disease, Beijing, 100069, China.
| |
Collapse
|
20
|
Shukla M, Abdul-Hay M, Choi JH. Molecular Features and Treatment Paradigms of Acute Myeloid Leukemia. Biomedicines 2024; 12:1768. [PMID: 39200232 PMCID: PMC11351617 DOI: 10.3390/biomedicines12081768] [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: 05/07/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 09/02/2024] Open
Abstract
Acute myeloid leukemia (AML) is a common hematologic malignancy that is considered to be a disease of aging, and traditionally has been treated with induction chemotherapy, followed by consolidation chemotherapy and/or allogenic hematopoietic stem cell transplantation. More recently, with the use of next-generation sequencing and access to molecular information, targeted molecular approaches to the treatment of AML have been adopted. Molecular targeting is gaining prominence, as AML mostly afflicts the elderly population, who often cannot tolerate traditional chemotherapy. Understanding molecular changes at the gene level is also important for accurate disease classification, risk stratification, and prognosis, allowing for more personalized medicine. Some mutations are well studied and have an established gene-specific therapy, including FLT3 and IDH1/2, while others are being investigated in clinical trials. However, data on most known mutations in AML are still minimal and therapeutic studies are in pre-clinical stages, highlighting the importance of further research and elucidation of the pathophysiology involving these genes. In this review, we aim to highlight the key molecular alterations and chromosomal changes that characterize AML, with a focus on pathophysiology, presently available treatment approaches, and future therapeutic options.
Collapse
Affiliation(s)
| | | | - Jun H. Choi
- Department of Hematology and Medical Oncology, NYU Langone Health, Perlmutter Cancer Center, New York, NY 10016, USA; (M.S.)
| |
Collapse
|
21
|
Sakthivel D, Brown-Suedel AN, Lopez KE, Salgar S, Coutinho LE, Keane F, Huang S, Sherry KM, Charendoff CI, Dunne KP, Robichaux DJ, Vargas-Hernández A, Le B, Shin CS, Carisey AF, Poreba M, Flanagan JM, Bouchier-Hayes L. Caspase-2 is essential for proliferation and self-renewal of nucleophosmin-mutated acute myeloid leukemia. SCIENCE ADVANCES 2024; 10:eadj3145. [PMID: 39093977 PMCID: PMC11296348 DOI: 10.1126/sciadv.adj3145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 06/28/2024] [Indexed: 08/04/2024]
Abstract
Mutation in nucleophosmin (NPM1) causes relocalization of this normally nucleolar protein to the cytoplasm (NPM1c+). Despite NPM1 mutation being the most common driver mutation in cytogenetically normal adult acute myeloid leukemia (AML), the mechanisms of NPM1c+-induced leukemogenesis remain unclear. Caspase-2 is a proapoptotic protein activated by NPM1 in the nucleolus. Here, we show that caspase-2 is also activated by NPM1c+ in the cytoplasm and DNA damage-induced apoptosis is caspase-2 dependent in NPM1c+ but not in NPM1wt AML cells. Strikingly, in NPM1c+ cells, caspase-2 loss results in profound cell cycle arrest, differentiation, and down-regulation of stem cell pathways that regulate pluripotency including impairment of the AKT/mTORC1 pathways, and inhibition of Rictor cleavage. In contrast, there were minimal differences in proliferation, differentiation, or the transcriptional profile of NPM1wt cells lacking caspase-2. Our results show that caspase-2 is essential for proliferation and self-renewal of AML cells expressing mutated NPM1. This study demonstrates that caspase-2 is a major effector of NPM1c+ function.
Collapse
Affiliation(s)
- Dharaniya Sakthivel
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexandra N. Brown-Suedel
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
| | - Karla E. Lopez
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
| | - Suruchi Salgar
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
| | - Luiza E. Coutinho
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
| | - Francesca Keane
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shixia Huang
- Advanced Technology Cores, Department of Molecular and Cellular Biology, Huffington Department of Education, Innovation & Technology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kenneth Mc Sherry
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chloé I. Charendoff
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kevin P. Dunne
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dexter J. Robichaux
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexander Vargas-Hernández
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
| | - BaoChau Le
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
| | - Crystal S. Shin
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexandre F. Carisey
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Marcin Poreba
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw 50370, Poland
| | - Jonathan M. Flanagan
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
| | - Lisa Bouchier-Hayes
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
22
|
Wu Q, Zhang Y, Yuan B, Huang Y, Jiang L, Liu F, Yan P, Cheng J, Long Z, Jiang X. Influence of genetic co-mutation on chemotherapeutic outcome in NPM1-mutated and FLT3-ITD wild-type AML patients. Cancer Med 2024; 13:e70102. [PMID: 39126219 PMCID: PMC11316012 DOI: 10.1002/cam4.70102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/27/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Nucleophosmin 1 (NPM1) gene-mutated acute myeloid leukemia (NPM1mut AML) is classified as a subtype with a favorable prognosis. However, some patients fail to achieve a complete remission or relapse after intensified chemotherapy. Genetic abnormalities in concomitant mutations contribute to heterogeneous prognosis of NPM1mut AML patients. METHODS In this study, 91 NPM1-mutated and FLT3-ITD wild-type (NPM1mut/FLT3-ITDwt) AML patients with intermediate-risk karyotype were enrolled to analyze the impact of common genetic co-mutations on chemotherapeutic outcome. RESULTS Our data revealed that TET1/2 (52/91, 57.1%) was the most prevalent co-mutation in NPM1mut AML patients, followed by IDH1/2 (36/91, 39.6%), DNMT3A (35/91, 38.5%), myelodysplastic syndrome related genes (MDS-related genes) (ASXL1, BCOR, EZH2, RUNX1, SF3B1, SRSF2, STAG2, U2AF1 and ZRSR2 genes) (35/91, 38.5%), FLT3-TKD (27/91, 29.7%) and GATA2 (13/91, 14.3%) mutations. Patients with TET1/2mut exhibited significantly worse relapse-free survival (RFS) (median, 28.7 vs. not reached (NR) months; p = 0.0382) compared to patients with TET1/2wt, while no significant difference was observed in overall survival (OS) (median, NR vs. NR; p = 0.3035). GATA2mut subtype was associated with inferior OS (median, 28 vs. NR months; p < 0.0010) and RFS (median, 24 vs. NR months; p = 0.0224) compared to GATA2wt. By multivariate analysis, GATA2mut and MDS-related genesmut were independently associated with worse survival. CONCLUSION Mutations in TET1/2, GATA2 and MDS-related genes were found to significantly influence the chemotherapeutic outcome of patients with NPM1mut AML. The findings of our study have significant clinical implications for identifying patients who have an adverse response to frontline chemotherapy and provide a novel reference for further prognostic stratification of NPM1mut/FLT3-ITDwt AML patients.
Collapse
Affiliation(s)
- Quan Wu
- Department of Hematology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Yujiao Zhang
- Department of Hematology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Baoyi Yuan
- Department of Hematology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Yun Huang
- Department of Hematology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Ling Jiang
- Department of Hematology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Fang Liu
- Department of Hematology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Ping Yan
- Department of Hematology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Jiaying Cheng
- Department of Hematology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Zhiquan Long
- Department of Hematology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Xuejie Jiang
- Department of Hematology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| |
Collapse
|
23
|
Taha MS, Ahmadian MR. Nucleophosmin: A Nucleolar Phosphoprotein Orchestrating Cellular Stress Responses. Cells 2024; 13:1266. [PMID: 39120297 PMCID: PMC11312075 DOI: 10.3390/cells13151266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Nucleophosmin (NPM1) is a key nucleolar protein released from the nucleolus in response to stress stimuli. NPM1 functions as a stress regulator with nucleic acid and protein chaperone activities, rapidly shuttling between the nucleus and cytoplasm. NPM1 is ubiquitously expressed in tissues and can be found in the nucleolus, nucleoplasm, cytoplasm, and extracellular environment. It plays a central role in various biological processes such as ribosome biogenesis, cell cycle regulation, cell proliferation, DNA damage repair, and apoptosis. In addition, it is highly expressed in cancer cells and solid tumors, and its mutation is a major cause of acute myeloid leukemia (AML). This review focuses on NPM1's structural features, functional diversity, subcellular distribution, and role in stress modulation.
Collapse
Affiliation(s)
- Mohamed S. Taha
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
- Research on Children with Special Needs Department, Institute of Medical Research and Clinical Studies, National Research Centre, Cairo 12622, Egypt
| | - Mohammad Reza Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| |
Collapse
|
24
|
Wei Z, Zhao Y, Cai J, Xie Y. The Nucleolar Protein C1orf131 Is a Novel Gene Involved in the Progression of Lung Adenocarcinoma Cells through the AKT Signalling Pathway. Int J Mol Sci 2024; 25:6381. [PMID: 38928092 PMCID: PMC11203618 DOI: 10.3390/ijms25126381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Lung adenocarcinoma (LUAD) is the most widespread cancer in the world, and its development is associated with complex biological mechanisms that are poorly understood. Here, we revealed a marked upregulation in the mRNA level of C1orf131 in LUAD samples compared to non-tumor tissue samples in The Cancer Genome Atlas (TCGA). Depletion of C1orf131 suppressed cell proliferation and growth, whereas it stimulated apoptosis in LUAD cells. Mechanistic investigations revealed that C1orf131 knockdown induced cell cycle dysregulation via the AKT and p53/p21 signalling pathways. Additionally, C1orf131 knockdown blocked cell migration through the modulation of epithelial-mesenchymal transition (EMT) in lung adenocarcinoma. Notably, we identified the C1orf131 protein nucleolar localization sequence, which included amino acid residues 137-142 (KKRKLT) and 240-245 (KKKRKG). Collectively, C1orf131 has potential as a novel therapeutic marker for patients in the future, as it plays a vital role in the progression of lung adenocarcinoma.
Collapse
Affiliation(s)
- Zhili Wei
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China;
| | - Yiming Zhao
- College of Medical Informatics, Chongqing Medical University, Chongqing 400016, China;
| | - Jing Cai
- National Talent Introduction Demonstration Base, the College of Basic Medicine, Harbin Medical University, Harbin 150081, China;
| | - Yajun Xie
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China;
| |
Collapse
|
25
|
Jang HJ, Min HY, Kang YP, Boo HJ, Kim J, Ahn JH, Oh SH, Jung JH, Park CS, Park JS, Kim SY, Lee HY. Tobacco-induced hyperglycemia promotes lung cancer progression via cancer cell-macrophage interaction through paracrine IGF2/IR/NPM1-driven PD-L1 expression. Nat Commun 2024; 15:4909. [PMID: 38851766 PMCID: PMC11162468 DOI: 10.1038/s41467-024-49199-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 05/23/2024] [Indexed: 06/10/2024] Open
Abstract
Tobacco smoking (TS) is implicated in lung cancer (LC) progression through the development of metabolic syndrome. However, direct evidence linking metabolic syndrome to TS-mediated LC progression remains to be established. Our findings demonstrate that 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone and benzo[a]pyrene (NNK and BaP; NB), components of tobacco smoke, induce metabolic syndrome characteristics, particularly hyperglycemia, promoting lung cancer progression in male C57BL/6 J mice. NB enhances glucose uptake in tumor-associated macrophages by increasing the expression and surface localization of glucose transporter (GLUT) 1 and 3, thereby leading to transcriptional upregulation of insulin-like growth factor 2 (IGF2), which subsequently activates insulin receptor (IR) in LC cells in a paracrine manner, promoting its nuclear import. Nuclear IR binds to nucleophosmin (NPM1), resulting in IR/NPM1-mediated activation of the CD274 promoter and expression of programmed death ligand-1 (PD-L1). Restricting glycolysis, depleting macrophages, or blocking PD-L1 inhibits NB-mediated LC progression. Analysis of patient tissues and public databases reveals elevated levels of IGF2 and GLUT1 in tumor-associated macrophages, as well as tumoral PD-L1 and phosphorylated insulin-like growth factor 1 receptor/insulin receptor (pIGF-1R/IR) expression, suggesting potential poor prognostic biomarkers for LC patients. Our data indicate that paracrine IGF2/IR/NPM1/PD-L1 signaling, facilitated by NB-induced dysregulation of glucose levels and metabolic reprogramming of macrophages, contributes to TS-mediated LC progression.
Collapse
Affiliation(s)
- Hyun-Ji Jang
- Creative Research Initiative Center for concurrent control of emphysema and lung cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hye-Young Min
- Creative Research Initiative Center for concurrent control of emphysema and lung cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yun Pyo Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hye-Jin Boo
- Creative Research Initiative Center for concurrent control of emphysema and lung cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Histology, College of Medicine, Jeju National University, Jeju, 63243, Republic of Korea
| | - Jisung Kim
- Creative Research Initiative Center for concurrent control of emphysema and lung cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jee Hwan Ahn
- Creative Research Initiative Center for concurrent control of emphysema and lung cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology and College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Ho Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jin Hwa Jung
- PET core, Convergence Medicine Research Center, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Choon-Sik Park
- Soonchunhyang University Bucheon Hospital, Bucheon-si, Gyeonggi-do, 14584, Republic of Korea
| | - Jong-Sook Park
- Soonchunhyang University Bucheon Hospital, Bucheon-si, Gyeonggi-do, 14584, Republic of Korea
| | - Seog-Young Kim
- PET core, Convergence Medicine Research Center, Asan Medical Center, Seoul, 05505, Republic of Korea
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Ho-Young Lee
- Creative Research Initiative Center for concurrent control of emphysema and lung cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
26
|
Algar S, Vázquez-Villa H, Aguilar-Garrido P, Navarro-Aguadero MÁ, Velasco-Estévez M, Sánchez-Merino A, Arribas-Álvarez I, Paradela A, Giner-Arroyo RL, Tamargo-Azpilicueta J, Díaz-Moreno I, Martínez-López J, Gallardo M, López-Rodríguez ML, Benhamú B. Cancer-Stem-Cell Phenotype-Guided Discovery of a Microbiota-Inspired Synthetic Compound Targeting NPM1 for Leukemia. JACS AU 2024; 4:1786-1800. [PMID: 38818079 PMCID: PMC11134387 DOI: 10.1021/jacsau.3c00682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 06/01/2024]
Abstract
The human microbiota plays an important role in human health and disease, through the secretion of metabolites that regulate key biological functions. We propose that microbiota metabolites represent an unexplored chemical space of small drug-like molecules in the search of new hits for drug discovery. Here, we describe the generation of a set of complex chemotypes inspired on selected microbiota metabolites, which have been synthesized using asymmetric organocatalytic reactions. Following a primary screening in CSC models, we identified the novel compound UCM-13369 (4b) whose cytotoxicity was mediated by NPM1. This protein is one of the most frequent mutations of AML, and NPM1-mutated AML is recognized by the WHO as a distinct hematopoietic malignancy. UCM-13369 inhibits NPM1 expression, downregulates the pathway associated with mutant NPM1 C+, and specifically recognizes the C-end DNA-binding domain of NPM1 C+, avoiding the nucleus-cytoplasm translocation involved in the AML tumorological process. The new NPM1 inhibitor triggers apoptosis in AML cell lines and primary cells from AML patients and reduces tumor infiltration in a mouse model of AML with NPM1 C+ mutation. The disclosed phenotype-guided discovery of UCM-13369, a novel small molecule inspired on microbiota metabolites, confirms that CSC death induced by NPM1 inhibition represents a promising therapeutic opportunity for NPM1-mutated AML, a high-mortality disease.
Collapse
Affiliation(s)
- Sergio Algar
- Department
of Organic Chemistry, Faculty of Chemistry, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Henar Vázquez-Villa
- Department
of Organic Chemistry, Faculty of Chemistry, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Pedro Aguilar-Garrido
- Department
of Haematology, Hospital Universitario 12
de Octubre, Instituto de Investigación Sanitaria Hospital 12
de Octubre (imas12), E-28041 Madrid, Spain
- H12O-CNIO
Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, E-28029 Madrid, Spain
| | - Miguel Ángel Navarro-Aguadero
- Department
of Haematology, Hospital Universitario 12
de Octubre, Instituto de Investigación Sanitaria Hospital 12
de Octubre (imas12), E-28041 Madrid, Spain
- H12O-CNIO
Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, E-28029 Madrid, Spain
| | - María Velasco-Estévez
- Department
of Haematology, Hospital Universitario 12
de Octubre, Instituto de Investigación Sanitaria Hospital 12
de Octubre (imas12), E-28041 Madrid, Spain
- H12O-CNIO
Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, E-28029 Madrid, Spain
| | - Anabel Sánchez-Merino
- Department
of Organic Chemistry, Faculty of Chemistry, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Iván Arribas-Álvarez
- Department
of Organic Chemistry, Faculty of Chemistry, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | | | - Rafael L. Giner-Arroyo
- Institute
for Chemical Research, cicCartuja, University
of Seville, CSIC, E-41092 Sevilla, Spain
| | | | - Irene Díaz-Moreno
- Institute
for Chemical Research, cicCartuja, University
of Seville, CSIC, E-41092 Sevilla, Spain
| | - Joaquín Martínez-López
- Department
of Haematology, Hospital Universitario 12
de Octubre, Instituto de Investigación Sanitaria Hospital 12
de Octubre (imas12), E-28041 Madrid, Spain
- H12O-CNIO
Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, E-28029 Madrid, Spain
| | - Miguel Gallardo
- Department
of Haematology, Hospital Universitario 12
de Octubre, Instituto de Investigación Sanitaria Hospital 12
de Octubre (imas12), E-28041 Madrid, Spain
- H12O-CNIO
Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, E-28029 Madrid, Spain
| | - María L. López-Rodríguez
- Department
of Organic Chemistry, Faculty of Chemistry, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Bellinda Benhamú
- Department
of Organic Chemistry, Faculty of Chemistry, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| |
Collapse
|
27
|
Bottardi S, Layne T, Ramòn AC, Quansah N, Wurtele H, Affar EB, Milot E. MNDA, a PYHIN factor involved in transcriptional regulation and apoptosis control in leukocytes. Front Immunol 2024; 15:1395035. [PMID: 38680493 PMCID: PMC11045911 DOI: 10.3389/fimmu.2024.1395035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 04/02/2024] [Indexed: 05/01/2024] Open
Abstract
Inflammation control is critical during the innate immune response. Such response is triggered by the detection of molecules originating from pathogens or damaged host cells by pattern-recognition receptors (PRRs). PRRs subsequently initiate intra-cellular signalling through different pathways, resulting in i) the production of inflammatory cytokines, including type I interferon (IFN), and ii) the initiation of a cascade of events that promote both immediate host responses as well as adaptive immune responses. All human PYRIN and HIN-200 domains (PYHIN) protein family members were initially proposed to be PRRs, although this view has been challenged by reports that revealed their impact on other cellular mechanisms. Of relevance here, the human PYHIN factor myeloid nuclear differentiation antigen (MNDA) has recently been shown to directly control the transcription of genes encoding factors that regulate programmed cell death and inflammation. While MNDA is mainly found in the nucleus of leukocytes of both myeloid (neutrophils and monocytes) and lymphoid (B-cell) origin, its subcellular localization has been shown to be modulated in response to genotoxic agents that induce apoptosis and by bacterial constituents, mediators of inflammation. Prior studies have noted the importance of MNDA as a marker for certain forms of lymphoma, and as a clinical prognostic factor for hematopoietic diseases characterized by defective regulation of apoptosis. Abnormal expression of MNDA has also been associated with altered levels of cytokines and other inflammatory mediators. Refining our comprehension of the regulatory mechanisms governing the expression of MNDA and other PYHIN proteins, as well as enhancing our definition of their molecular functions, could significantly influence the management and treatment strategies of numerous human diseases. Here, we review the current state of knowledge regarding PYHIN proteins and their role in innate and adaptive immune responses. Emphasis will be placed on the regulation, function, and relevance of MNDA expression in the control of gene transcription and RNA stability during cell death and inflammation.
Collapse
Affiliation(s)
- Stefania Bottardi
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Est-de-l’Île de Montreal, Montreal, QC, Canada
| | - Taylorjade Layne
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Est-de-l’Île de Montreal, Montreal, QC, Canada
| | - Ailyn C. Ramòn
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Est-de-l’Île de Montreal, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Norreen Quansah
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Est-de-l’Île de Montreal, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Hugo Wurtele
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Est-de-l’Île de Montreal, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - El Bachir Affar
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Est-de-l’Île de Montreal, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Eric Milot
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Est-de-l’Île de Montreal, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
28
|
Balčiūnienė E, Inčiūra A, Juozaitytė E, Ugenskienė R. Impact of RRP1B Variants on the Phenotype, Progression, and Metastasis of Cervical Cancer. Cancers (Basel) 2024; 16:1250. [PMID: 38610928 PMCID: PMC11011178 DOI: 10.3390/cancers16071250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/17/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Metastasis is a key determinant of cancer progression, influenced significantly by genetic mechanisms. RRP1B, primarily a nucleolar protein, emerges as a suppressor of metastasis, forming alliances with various cellular components and modulating gene expression. This study investigates the involvement of the ribosomal RNA processing 1 homolog B (RRP1B) gene in metastasis regulation in cervical cancer. Through a comprehensive analysis of 172 cervical cancer patients, we evaluated five RRP1B single nucleotide polymorphisms (SNPs) (rs2838342, rs7276633, rs2051407, rs9306160, and rs762400) for their associations with clinicopathological features and survival outcomes. Significant associations were observed between specific genetic variants and clinicopathological parameters. Notably, the A allele of rs2838342 was associated with reduced odds of advanced tumor size, worse prognosis, and, preliminarily, distant metastasis, while the T allele of rs7276633 correlated with a decreased risk of higher tumor size and worse prognosis. Additionally, the C allele of rs2051407 demonstrated protective effects against larger tumors, metastasis, and adverse prognosis. The rs9306160 C allele exhibited a protective effect against metastasis. The rs762400 G allele was significant for reduced tumor size and metastasis risk. Furthermore, the rs2838342 A allele, rs7276633 T allele, rs2051407 C allele, and rs762400 G allele were associated with improved overall survival, demonstrating their potential significance in predicting prognoses in cervical cancer. Linkage disequilibrium and haplotypes analysis enabled us to evaluate the collective effect of the analyzed SNPs, which was in line with the results of allelic models. Our findings underscore the clinical relevance of RRP1B SNPs as prognostic markers in cervical cancer, shedding light on the intricate interplay between genetic factors and disease-progression dynamics. This research provides critical insights for future investigations and underscores the importance of incorporating RRP1B SNP detection into prognostic-assessment tools for accurate prediction of disease outcomes in cervical cancer.
Collapse
Affiliation(s)
- Eglė Balčiūnienė
- Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (A.I.); (E.J.); (R.U.)
| | - Arturas Inčiūra
- Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (A.I.); (E.J.); (R.U.)
| | - Elona Juozaitytė
- Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (A.I.); (E.J.); (R.U.)
| | - Rasa Ugenskienė
- Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (A.I.); (E.J.); (R.U.)
- Department of Genetics and Molecular Medicine, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| |
Collapse
|
29
|
Khan I, Amin MA, Eklund EA, Gartel AL. Regulation of HOX gene expression in AML. Blood Cancer J 2024; 14:42. [PMID: 38453907 PMCID: PMC10920644 DOI: 10.1038/s41408-024-01004-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/12/2024] [Accepted: 02/14/2024] [Indexed: 03/09/2024] Open
Abstract
As key developmental regulators, HOX cluster genes have varied and context-specific roles in normal and malignant hematopoiesis. A complex interaction of transcription factors, epigenetic regulators, long non-coding RNAs and chromatin structural changes orchestrate HOX expression in leukemia cells. In this review we summarize molecular mechanisms underlying HOX regulation in clinical subsets of AML, with a focus on NPM1 mutated (NPM1mut) AML comprising a third of all AML patients. While the leukemia initiating function of the NPM1 mutation is clearly dependent on HOX activity, the favorable treatment responses in these patients with upregulation of HOX cluster genes is a poorly understood paradoxical observation. Recent data confirm FOXM1 as a suppressor of HOX activity and a well-known binding partner of NPM suggesting that FOXM1 inactivation may mediate the effect of cytoplasmic NPM on HOX upregulation. Conversely the residual nuclear fraction of mutant NPM has also been recently shown to have chromatin modifying effects permissive to HOX expression. Recent identification of the menin-MLL interaction as a critical vulnerability of HOX-dependent AML has fueled the development of menin inhibitors that are clinically active in NPM1 and MLL rearranged AML despite inconsistent suppression of the HOX locus. Insights into context-specific regulation of HOX in AML may provide a solid foundation for targeting this common vulnerability across several major AML subtypes.
Collapse
Affiliation(s)
- Irum Khan
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
- Department of Medicine at the Feinberg School of Medicine, Northwestern University, Chicago, USA
| | - Mohammed A Amin
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Elizabeth A Eklund
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
- Department of Medicine at the Feinberg School of Medicine, Northwestern University, Chicago, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Andrei L Gartel
- Department of Medicine, University of Illinois, Chicago, IL, USA.
| |
Collapse
|
30
|
Ruiz M, Jindal K, Casey V, Soares LM, Manuguid F, Moehler T. Uptake of novel therapies into first-line treatment for acute myeloid leukemia patients: EU4 + UK perspective. Future Oncol 2024; 20:533-546. [PMID: 37975244 DOI: 10.2217/fon-2022-1178] [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] [Indexed: 11/19/2023] Open
Abstract
Aim: To explore the incorporation of novel agents in the first-line setting for acute myeloid leukemia patients. Materials & methods: Observational study based on data from a multi-country cross-sectional retrospective web-based survey sent to 518 physicians in Europe between 2020 and 2021. Information from 2040 patients was analyzed. Results: 604 patients (29.6%) received novel agents in both intensive and non-intensive setting. Comorbidities were not a barrier for the use of novel agents. The presence of tumor mutations was observed to be an important element for treatment decision. Conclusion: There is a progressive incorporation of novel agents for newly diagnosed acute myeloid leukemia patients.
Collapse
Affiliation(s)
- Magdalena Ruiz
- Hemato-Oncology Data Sciences, Safety & Medical, IQVIA Madrid & Frankfurt
| | - Kriti Jindal
- Global Oncology, Real-World & Analytics Solutions, IQVIA London
| | - Vicky Casey
- Data Science & Advanced Analytics, IQVIA London
| | | | | | - Thomas Moehler
- Hemato-Oncology Data Sciences, Safety & Medical, IQVIA Madrid & Frankfurt
| |
Collapse
|
31
|
Tang S, Wang Y, Luo R, Fang R, Liu Y, Xiang H, Ran P, Tong Y, Sun M, Tan S, Huang W, Huang J, Lv J, Xu N, Yao Z, Zhang Q, Xu Z, Yue X, Yu Z, Akesu S, Ding Y, Xu C, Lu W, Zhou Y, Hou Y, Ding C. Proteomic characterization identifies clinically relevant subgroups of soft tissue sarcoma. Nat Commun 2024; 15:1381. [PMID: 38360860 PMCID: PMC10869728 DOI: 10.1038/s41467-024-45306-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 01/18/2024] [Indexed: 02/17/2024] Open
Abstract
Soft tissue sarcoma is a broad family of mesenchymal malignancies exhibiting remarkable histological diversity. We portray the proteomic landscape of 272 soft tissue sarcomas representing 12 major subtypes. Hierarchical classification finds the similarity of proteomic features between angiosarcoma and epithelial sarcoma, and elevated expression of SHC1 in AS and ES is correlated with poor prognosis. Moreover, proteomic clustering classifies patients of soft tissue sarcoma into 3 proteomic clusters with diverse driven pathways and clinical outcomes. In the proteomic cluster featured with the high cell proliferation rate, APEX1 and NPM1 are found to promote cell proliferation and drive the progression of cancer cells. The classification based on immune signatures defines three immune subtypes with distinctive tumor microenvironments. Further analysis illustrates the potential association between immune evasion markers (PD-L1 and CD80) and tumor metastasis in soft tissue sarcoma. Overall, this analysis uncovers sarcoma-type-specific changes in proteins, providing insights about relationships of soft tissue sarcoma.
Collapse
Affiliation(s)
- Shaoshuai Tang
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences, Human Phenome Institute, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Yunzhi Wang
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences, Human Phenome Institute, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Rongkui Luo
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rundong Fang
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences, Human Phenome Institute, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Yufeng Liu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hang Xiang
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences, Human Phenome Institute, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Peng Ran
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences, Human Phenome Institute, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Yexin Tong
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences, Human Phenome Institute, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Mingjun Sun
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences, Human Phenome Institute, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Subei Tan
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences, Human Phenome Institute, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Wen Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jie Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiacheng Lv
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences, Human Phenome Institute, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Ning Xu
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences, Human Phenome Institute, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Zhenmei Yao
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences, Human Phenome Institute, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Qiao Zhang
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences, Human Phenome Institute, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Ziyan Xu
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences, Human Phenome Institute, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Xuetong Yue
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences, Human Phenome Institute, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Zixiang Yu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Sujie Akesu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuqin Ding
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Medical Imaging, Shanghai, China
| | - Chen Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Weiqi Lu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Yuhong Zhou
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Chen Ding
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences, Human Phenome Institute, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
32
|
Castaño-Díez S, Guijarro F, López-Guerra M, Pérez-Valencia AI, Gómez-Núñez M, Colomer D, Díaz-Beyá M, Esteve J, Rozman M. Infrequent Presentations of Chronic NPM1-Mutated Myeloid Neoplasms: Clinicopathological Features of Eight Cases from a Single Institution and Review of the Literature. Cancers (Basel) 2024; 16:705. [PMID: 38398096 PMCID: PMC10886643 DOI: 10.3390/cancers16040705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/18/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Non-acute myeloid neoplasms (MNs) with NPM1 mutations (NPM1mut-MNs) pose a diagnostic and therapeutic dilemma, primarily manifesting as chronic myelomonocytic leukemia (CMML) and myelodysplastic syndromes (MDS). The classification and treatment approach for these conditions as acute myeloid leukemia (AML) are debated. We describe eight cases of atypical NPM1mut-MNs from our institution and review the literature. We include a rare case of concurrent prostate carcinoma and MN consistent with chronic eosinophilic leukemia, progressing to myeloid sarcoma of the skin. Of the remaining seven cases, five were CMML and two were MDS. NPM1 mutations occur in 3-5% of CMML and 1-6% of MDS, with an increased likelihood of rapid evolution to AML. Their influence on disease progression varies, and their prognostic significance in non-acute MNs is less established than in AML. Non-acute MNs with NPM1 mutations may display an aggressive clinical course, emphasizing the need for a comprehensive diagnosis integrating clinical and biological data. Tailoring patient management on an individualized basis, favoring intensive treatment aligned with AML protocols, is crucial, regardless of blast percentage. Research on the impact of NPM1 mutations in non-acute myeloid neoplasms is ongoing, requiring challenging prospective studies with substantial patient cohorts and extended follow-up periods for validation.
Collapse
Affiliation(s)
- Sandra Castaño-Díez
- Hematology Department, Hospital Clínic Barcelona, 08036 Barcelona, Spain; (S.C.-D.); (A.I.P.-V.); (M.D.-B.); (J.E.)
- Medical School, University of Barcelona, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (F.G.); (M.L.-G.); (D.C.)
| | - Francesca Guijarro
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (F.G.); (M.L.-G.); (D.C.)
- Hematopathology Section, Servei d’Anatomia Patològica, CDB, Hospital Clínic Barcelona, 08036 Barcelona, Spain
| | - Mònica López-Guerra
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (F.G.); (M.L.-G.); (D.C.)
- Hematopathology Section, Servei d’Anatomia Patològica, CDB, Hospital Clínic Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Amanda Isabel Pérez-Valencia
- Hematology Department, Hospital Clínic Barcelona, 08036 Barcelona, Spain; (S.C.-D.); (A.I.P.-V.); (M.D.-B.); (J.E.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (F.G.); (M.L.-G.); (D.C.)
| | | | - Dolors Colomer
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (F.G.); (M.L.-G.); (D.C.)
- Hematopathology Section, Servei d’Anatomia Patològica, CDB, Hospital Clínic Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Marina Díaz-Beyá
- Hematology Department, Hospital Clínic Barcelona, 08036 Barcelona, Spain; (S.C.-D.); (A.I.P.-V.); (M.D.-B.); (J.E.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (F.G.); (M.L.-G.); (D.C.)
- Josep Carreras Leukemia Research Institute, 08916 Badalona, Spain
| | - Jordi Esteve
- Hematology Department, Hospital Clínic Barcelona, 08036 Barcelona, Spain; (S.C.-D.); (A.I.P.-V.); (M.D.-B.); (J.E.)
- Medical School, University of Barcelona, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (F.G.); (M.L.-G.); (D.C.)
- Josep Carreras Leukemia Research Institute, 08916 Badalona, Spain
| | - María Rozman
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (F.G.); (M.L.-G.); (D.C.)
- Hematopathology Section, Servei d’Anatomia Patològica, CDB, Hospital Clínic Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
33
|
Wang H, Zhou Z, Zhang J, Hao T, Wang P, Wu P, Su R, Yang H, Deng G, Chen S, Gu L, He Y, Zeng L, Zhang C, Yin S. Pumilio1 regulates NPM3/NPM1 axis to promote PD-L1-mediated immune escape in gastric cancer. Cancer Lett 2024; 581:216498. [PMID: 38029539 DOI: 10.1016/j.canlet.2023.216498] [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: 09/25/2023] [Revised: 11/08/2023] [Accepted: 11/16/2023] [Indexed: 12/01/2023]
Abstract
Abnormal regulation of RNA binding proteins (RBPs) plays an essential role in tumorigenesis and progression, but their functions and mechanisms remain largely elusive. Previously, we reported that Pumilio 1 (PUM1), a RBP, could regulate glycolysis metabolism and promote the progression of gastric cancer (GC). However, the role of PUM1 in tumor immune regulation remains largely elusive. In this study, we report that PUM1 induces immune escape through posttranscriptional regulation of PD-L1 in GC. We used multiplexed immunohistochemistry to analyze the correlation between PUM1 expression and immune microenvironment in GC. The effect of PUM1 deficiency on tumor killing of T cells was examined in vitro and in vivo. The molecular mechanism of PUM1 was evaluated via RNA immunoprecipitation, chromatin immunoprecipitation, Western blot, co-immunoprecipitation, and RNA stability assays. Clinically, elevated PUM1 expression is associated with high-expression of PD-L1, lack of CD8+ T cell infiltration and poor prognosis in GC patients. PUM1 positively regulates PD-L1 expression and PUM1 reduction enhances T cell killing of tumors. Mechanistically, PUM1 directly binds to nucleophosmin/nucleoplasmin 3 (NPM3) mRNA and stabilizes NPM3. NPM3 interacts with NPM1 to promote NPM1 translocation into the nucleus and increase the transcription of PD-L1. PUM1 inhibits the anti-tumor activity of T cells through the PUM1/NPM3/PD-L1 axis. In summary, this study reveals the critical post-transcriptional effect of PUM1 in the modulation of PD-L1-dependent GC immune escape, thus provides a novel indicator and potential therapeutic target for cancer immunotherapy.
Collapse
Affiliation(s)
- Han Wang
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China; Department of Gastrointestinal Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Zhijun Zhou
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Junchang Zhang
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China; Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Tengfei Hao
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Pengliang Wang
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Pei Wu
- Department of Gastrointestinal Surgery, Yongchuan Hospital of Chongqing Medical university, Chongqing, China
| | - Rishun Su
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Huan Yang
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Guofei Deng
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Songyao Chen
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Liang Gu
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yulong He
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China; Department of Gastrointestinal Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Leli Zeng
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China.
| | - Changhua Zhang
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China.
| | - Songcheng Yin
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China.
| |
Collapse
|
34
|
Abramov DS, Fedorova AS, Tuzova EA, Myakova NV, Konovalov DM. [ALK-positive anaplastic large cell lymphoma of paranasal sinuses: two cases report and literature review]. Arkh Patol 2024; 86:42-47. [PMID: 39073541 DOI: 10.17116/patol20248604142] [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] [Indexed: 07/30/2024]
Abstract
ALK-positive anaplastic large cell lymphoma is a rare T-cell lymphoma with ALK gene rearrangement that develops in children and young adults. The disease almost always affects the lymph nodes, and extranodal areas are also frequently involved. This article describes two cases of atypical localization of ALK-positive anaplastic large cell lymphoma with involvement of the paranasal sinuses.
Collapse
Affiliation(s)
- D S Abramov
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - A S Fedorova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - E A Tuzova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - N V Myakova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - D M Konovalov
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
- Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| |
Collapse
|
35
|
Mende H, Khatri A, Lange C, Poveda-Cuevas SA, Tascher G, Covarrubias-Pinto A, Löhr F, Koschade SE, Dikic I, Münch C, Bremm A, Brunetti L, Brandts CH, Uckelmann H, Dötsch V, Rogov VV, Bhaskara RM, Müller S. An atypical GABARAP binding module drives the pro-autophagic potential of the AML-associated NPM1c variant. Cell Rep 2023; 42:113484. [PMID: 37999976 DOI: 10.1016/j.celrep.2023.113484] [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: 03/29/2023] [Revised: 09/22/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
The nucleolar scaffold protein NPM1 is a multifunctional regulator of cellular homeostasis, genome integrity, and stress response. NPM1 mutations, known as NPM1c variants promoting its aberrant cytoplasmic localization, are the most frequent genetic alterations in acute myeloid leukemia (AML). A hallmark of AML cells is their dependency on elevated autophagic flux. Here, we show that NPM1 and NPM1c induce the autophagy-lysosome pathway by activating the master transcription factor TFEB, thereby coordinating the expression of lysosomal proteins and autophagy regulators. Importantly, both NPM1 and NPM1c bind to autophagy modifiers of the GABARAP subfamily through an atypical binding module preserved within its N terminus. The propensity of NPM1c to induce autophagy depends on this module, likely indicating that NPM1c exerts its pro-autophagic activity by direct engagement with GABARAPL1. Our data report a non-canonical binding mode of GABARAP family members that drives the pro-autophagic potential of NPM1c, potentially enabling therapeutic options.
Collapse
Affiliation(s)
- Hannah Mende
- Goethe University Frankfurt, Institute of Biochemistry II, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Anshu Khatri
- Goethe University Frankfurt, Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Max-von-Laue Street 9, 60438 Frankfurt, Germany
| | - Carolin Lange
- Goethe University Frankfurt, Institute of Biochemistry II, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany; Goethe University Frankfurt, Buchmann Institute for Molecular Life Sciences, Max-von-Laue Street 15, 60438 Frankfurt, Germany
| | - Sergio Alejandro Poveda-Cuevas
- Goethe University Frankfurt, Institute of Biochemistry II, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany; Goethe University Frankfurt, Buchmann Institute for Molecular Life Sciences, Max-von-Laue Street 15, 60438 Frankfurt, Germany
| | - Georg Tascher
- Goethe University Frankfurt, Institute of Biochemistry II, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Adriana Covarrubias-Pinto
- Goethe University Frankfurt, Institute of Biochemistry II, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Frank Löhr
- Goethe University Frankfurt, Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Max-von-Laue Street 9, 60438 Frankfurt, Germany
| | - Sebastian E Koschade
- Goethe University Frankfurt, Institute of Biochemistry II, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany; Goethe University Frankfurt, University Hospital, Department of Medicine, Hematology/Oncology, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Ivan Dikic
- Goethe University Frankfurt, Institute of Biochemistry II, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Christian Münch
- Goethe University Frankfurt, Institute of Biochemistry II, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Anja Bremm
- Goethe University Frankfurt, Institute of Biochemistry II, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Lorenzo Brunetti
- Marche Polytechnic University, Department of Clinical and Molecular Sciences, Via Tronto 10, 60020 Ancona, Italy
| | - Christian H Brandts
- Goethe University Frankfurt, University Hospital, Department of Medicine, Hematology/Oncology, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Hannah Uckelmann
- Goethe University Frankfurt, University Hospital, Department of Pediatrics, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Volker Dötsch
- Goethe University Frankfurt, Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Max-von-Laue Street 9, 60438 Frankfurt, Germany
| | - Vladimir V Rogov
- Goethe University Frankfurt, Institute of Pharmaceutical Chemistry, Max-von-Laue Street 15, 60438 Frankfurt, Germany; Goethe University Frankfurt, Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Max-von-Laue Street 15, 60438 Frankfurt, Germany
| | - Ramachandra M Bhaskara
- Goethe University Frankfurt, Institute of Biochemistry II, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany; Goethe University Frankfurt, Buchmann Institute for Molecular Life Sciences, Max-von-Laue Street 15, 60438 Frankfurt, Germany.
| | - Stefan Müller
- Goethe University Frankfurt, Institute of Biochemistry II, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
| |
Collapse
|
36
|
Liu Y, Zhang L, Chen X, Sun C, Zhang Y, Li Y, Li C. Functional characterization of porcine nucleophosmin (NPM1) gene in promoting the replication of Japanese encephalitis virus and induction of inflammatory cytokines. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 148:104902. [PMID: 37536401 DOI: 10.1016/j.dci.2023.104902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Nucleophosmin (NPM1) is a multifunctional nucleolar protein that plays a role in cell cycle control, tumorigenesis, induction of the inflammatory cytokine, virus replication, as well as the cellular responses to a variety of stress stimuli. However, its physiological functions in pigs have not been well understood. Here, we cloned the porcine NPM1 (porNPM1) gene and analyzed the functions of the porNPM1 protein in pigs. The full-length porNPM1 gene encoded a 294-amino acid protein with 94.5%-99.3% sequence identity to its orthologues in mammals and was extensively expressed in various pig tissues at the mRNA level. The porNPM1 primarily localizes in the nucleus of ST cells, while it translocates from the nucleus to nucleoplasm upon UV irradiation or H2O2 treatment. Notably, JEV infection blocked the translocation of porNPM1 from the nucleolus to the nucleoplasm. Furthermore, porNPM1 interacted with the JEV C protein and facilitated JEV replication in ST cells. The overexpression and knockdown of porNPM1 respectively enhanced or impaired JEV replication, suggesting the important role of porNPM1 in JEV replication. Additionally, the purified ectodomain of porNPM1 induced the production of inflammatory cytokines (TNF-α, IL-6, and IL-8). Together, these data demonstrated that porNPM1 is involved in cellular stress stimuli, JEV replication, and induction of inflammatory cytokines.
Collapse
Affiliation(s)
- Ying Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Linjie Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Xuan Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Chuwen Sun
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Yanbing Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi, 832003, China
| | - Yanhua Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; Comparative Medicine Research Institute, Yangzhou University, Yangzhou, 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China.
| | - Chenxi Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; Comparative Medicine Research Institute, Yangzhou University, Yangzhou, 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China.
| |
Collapse
|
37
|
Wu PS, Wang CY, Hsu HJ, Yen JH, Wu MJ. 8-Hydroxydaidzein Induces Apoptosis and Inhibits AML-Associated Gene Expression in U-937 Cells: Potential Phytochemical for AML Treatment. Biomolecules 2023; 13:1575. [PMID: 38002257 PMCID: PMC10669020 DOI: 10.3390/biom13111575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 09/30/2023] [Accepted: 10/19/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND 8-hydroxydaidzein (8-OHD) is a compound derived from daidzein, known for its anti-inflammatory and anti-proliferative properties in K562 human chronic myeloid leukemia (CML) cells. However, its effects on acute myeloid leukemia (AML) cells have not been fully understood. METHOD To investigate its potential anti-AML mechanism, we employed an integrated in vitro-in silico approach. RESULTS Our findings demonstrate that 8-OHD suppresses the expression of CDK6 and CCND2 proteins and induces cell apoptosis in U-937 cells by activating Caspase-7 and cleaving PARP-1. Microarray analysis revealed that 8-OHD downregulates differentially expressed genes (DEGs) associated with rRNA processing and ribosome biogenesis pathways. Moreover, AML-target genes, including CCND2, MYC, NPM1, FLT3, and TERT, were downregulated by 8-OHD. Additionally, molecular docking software predicted that 8-OHD has the potential to interact with CDK6, FLT3, and TERT proteins, thereby reducing their activity and inhibiting cell proliferation. Notably, we discovered a synergic pharmacological interaction between 8-OHD and cytarabine (Ara-C). CONCLUSIONS Overall, this study provides insights into the therapeutic applications of 8-OHD in treating AML and elucidates its underlying mechanisms of action.
Collapse
Affiliation(s)
- Pei-Shan Wu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan 717301, Taiwan;
- Department of Biotechnology, Chia Nan University of Pharmacy and Science, Tainan 717301, Taiwan
| | - Chih-Yang Wang
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, Taipei Medical University, Taipei 110301, Taiwan;
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei 110301, Taiwan
| | - Hao-Jen Hsu
- Department of Biomedical Sciences and Engineering, Tzu Chi University, Hualien 970, Taiwan;
| | - Jui-Hung Yen
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 970374, Taiwan;
- Institute of Medical Sciences, Tzu Chi University, Hualien 970374, Taiwan
| | - Ming-Jiuan Wu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan 717301, Taiwan;
- Department of Biotechnology, Chia Nan University of Pharmacy and Science, Tainan 717301, Taiwan
| |
Collapse
|
38
|
Hong Z, Xu C, Zheng S, Wang X, Tao Y, Tan Y, Lin G, Wu D, Ye D. Nucleophosmin 1 cooperates with BRD4 to facilitate c-Myc transcription to promote prostate cancer progression. Cell Death Discov 2023; 9:392. [PMID: 37875480 PMCID: PMC10597990 DOI: 10.1038/s41420-023-01682-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 10/08/2023] [Accepted: 10/12/2023] [Indexed: 10/26/2023] Open
Abstract
Nucleophosmin 1 (NPM1) is a multifunctional protein that promotes tumor progression in various cancers and is associated with a poor prognosis of prostate cancer (PCa). However, the mechanism by which NPM1 exerts its malignant potential in PCa remains elusive. Here, we showed that NPM1 is overexpressed in PCa cell lines and tissues and that the dysregulation of NPM1 promotes PCa proliferation. We also demonstrated that NPM1 transcriptionally upregulates c-Myc expression in PCa cells that is diminished by blockade of bromodomain-containing protein 4 (BRD4). Furthermore, we detected a correlation between NPM1 and c-Myc in patient PCa specimens. Mechanistically, NPM1 influences and cooperates with BRD4 to facilitate c-Myc transcription to promote PCa progression. In addition, JQ1, a bromodomain and extra-terminal domain (BET) inhibitor, in combination with NPM1 inhibition suppresses PCa progression in vitro and in vivo. These results indicate that NPM1 promotes PCa progression through a c-Myc -mediated pathway via BRD4, and blockade of the NPM1-c-Myc oncogenic pathway may be a therapeutic strategy for PCa.
Collapse
Affiliation(s)
- Zhe Hong
- Department of Urology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Shanghai Genitourinary Cancer Institute, 200032, Shanghai, China
| | - Chengdang Xu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, 200065, Shanghai, China
| | - Shengfeng Zheng
- Department of Urology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Shanghai Genitourinary Cancer Institute, 200032, Shanghai, China
| | - Xinan Wang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, 200065, Shanghai, China
| | - Yiran Tao
- Department of Urology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 317000, Taizhou, China
| | - Yao Tan
- Department of Urology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
| | - Guowen Lin
- Department of Urology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Shanghai Genitourinary Cancer Institute, 200032, Shanghai, China
| | - Denglong Wu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, 200065, Shanghai, China.
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China.
- Shanghai Genitourinary Cancer Institute, 200032, Shanghai, China.
| |
Collapse
|
39
|
Wei Q, Zhou J, Wang X, Li Z, Chen X, Chen K, Jiang R. Pan-cancer analysis of the prognostic and immunological role of nucleophosmin/nucleoplasmin 3 ( NPM3) and its potential significance in lung adenocarcinoma. CANCER PATHOGENESIS AND THERAPY 2023; 1:238-252. [PMID: 38327603 PMCID: PMC10846304 DOI: 10.1016/j.cpt.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/23/2023] [Accepted: 06/30/2023] [Indexed: 02/09/2024]
Abstract
Background Nucleophosmin/nucleoplasmin 3 (NPM3), a member of the NPM protein family, is widely expressed in various human tissues. Although previous studies identified elevated NPM3 expression in several cancers, a systematic pan-cancer analysis remains lacking. In this study, we conducted a comprehensive analysis of NPM3 to determine its role in tumorigenesis and tumor development. Methods Using data from The Cancer Genome Atlas (TCGA) and various bioinformatics analysis tools, we conducted a pan-cancer analysis of NPM3. Additionally, we collected gene expression and clinical data from 890 patients with lung adenocarcinoma (LUAD) from TCGA and the Gene Expression Omnibus database. We performed Cox regression analyses to explore the independent prognostic value of NPM3 expression in LUAD and plotted a nomogram to predict patient survival. We also used real-time quantitative polymerase chain reaction (RT-qPCR) to examine the expression levels of NPM3 in seven pairs of LUAD and paraneoplastic tissue samples. Results NPM3 expression was significantly increased in 20 types of cancer and was associated with poor prognosis in five types (P < 0.05). NPM3 expression was negatively correlated with DNA methylation and positively correlated with copy number variation. NPM3 was also significantly associated with immune cell infiltration in various cancers. Cox regression analyses revealed that NPM3 expression could serve as an independent prognostic marker of LUAD. Moreover, our nomogram demonstrated good predictive ability for the prognosis of patients with LUAD. Finally, the high expression of NPM3 in LUAD was verified using RT-qPCR. Conclusion NPM3 is a promising biomarker for predicting pan-cancer prognosis and immunotherapeutic efficacy.
Collapse
Affiliation(s)
- Qianhui Wei
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300202, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300202, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300202, China
- Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin 300202, China
| | - Jing Zhou
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300202, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300202, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300202, China
- Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin 300202, China
| | - Xinyue Wang
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300202, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300202, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300202, China
- Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin 300202, China
| | - Zhaona Li
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300202, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300202, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300202, China
- Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin 300202, China
| | - Xiuqiong Chen
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300202, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300202, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300202, China
- Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin 300202, China
| | - Kaidi Chen
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300202, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300202, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300202, China
- Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin 300202, China
| | - Richeng Jiang
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300202, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300202, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300202, China
- Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin 300202, China
| |
Collapse
|
40
|
Kaseb H, Visconte V, Socha DS, Crane GM, Durkin L, Cook JR, Maciejewski JP, Hsi ED, Rogers HJ. The clinicopathologic significance of NPM1 mutation and ability to detect mutated NPM1 by immunohistochemistry in non-AML myeloid neoplasms. Genes Chromosomes Cancer 2023; 62:573-580. [PMID: 36959701 PMCID: PMC11104021 DOI: 10.1002/gcc.23139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023] Open
Abstract
NPM1 mutated non-AML myeloid neoplasms (MN; <20% blasts) are characterized by an aggressive clinical course in a few studies. In this retrospective study, we evaluate the clinicopathologic and immunohistochemical features of non-AML MN patients with NPM1 mutations. We assessed NPM1 mutation by targeted next generation sequencing (NGS). Cytoplasmic NPM1 expression was assessed by immunohistochemistry (IHC) on formalin-fixed, formic acid-decalcified bone marrow biopsy specimens. We evaluated 34 non-AML MN patients with NPM1 mutations comprising MDS (22), MPN (3) and MDS/MPN (9). They commonly presented with anemia (88%), thrombocytopenia (58%) and leukopenia (50%). Bone marrow dysplasia was common (79%). The karyotype was often normal (64%). NGS for MN-associated mutations performed in a subset of the patients showed a median of 3 mutations. NPM1 mutations were more often missense (c.859C > T p. L287F; 65%) than frameshift insertion/duplication (35%) with median variant allele frequency (VAF; 9.7%, range 5.1%-49.8%). Mutated NPM1 by IHC showed cytoplasmic positivity in 48% and positivity was associated with higher VAF. The median overall survival (OS) in this cohort was 70 months. Nine patients (26%) progressed to AML. OS in patients who progressed to AML was significantly shorter than the one of patients without progression to AML (OS 20 vs. 128 months, respectively, log rank p = 0.05). NPM1 mutated non-AML MN patients commonly had cytopenias, dysplasia, normal karyotype, mutations in multiple genes, and an unfavorable clinical outcome, including progression to AML. Our data demonstrated that IHC for NPM1 can be a useful supplementary tool to predict NPM1 mutation in some non-AML MN; however, genetic testing cannot be replaced by IHC assessment.
Collapse
Affiliation(s)
- Hatem Kaseb
- Department of Pathology, University of Central Florida College of Medicine, Orlando, Florida, USA
| | - Valeria Visconte
- Department of Translational Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Daniel S. Socha
- Department of Pathology, OhioHealth, Riverside Methodist Hospital, Columbus, Ohio, USA
| | - Genevieve M. Crane
- Department of Laboratory Medicine, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Lisa Durkin
- Department of Laboratory Medicine, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - James R. Cook
- Department of Laboratory Medicine, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jaroslaw P. Maciejewski
- Department of Translational Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Hematologic Oncology and Blood Disorders, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Eric D. Hsi
- Department of Pathology and Laboratory Medicine, Wake Forest School of Medicine, Wake Forest Baptist Health, Winston-Salem, North Carolina, USA
| | - Heesun J. Rogers
- Department of Laboratory Medicine, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
41
|
Wolfová K, Otevřelová P, Holoubek A, Brodská B. Nucleolar phosphoprotein modifications as a marker of apoptosis induced by RITA treatment. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119501. [PMID: 37276927 DOI: 10.1016/j.bbamcr.2023.119501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/19/2023] [Accepted: 05/21/2023] [Indexed: 06/07/2023]
Abstract
Reactivating p53 and Inducing Tumor Apoptosis (RITA) has been reported to increase the p53 activity and to trigger p53-dependent apoptosis in cancer cells with wild-type p53. Tumor suppressor p53 interacts with nucleolar phosphoproteins nucleophosmin (NPM) and nucleolin (NCL), which have crucial role in many cellular processes. Specific NPM mutations associated with acute myeloid leukemia (AML) cause aberrant localization of NPM and p53 in the cytoplasm with possible impact on the p53 function. We tested an effect of RITA on primary cells, and we found significant RITA-induced changes in NPM and NCL phosphorylation associated with apoptosis in cells of AML patients, but not that of healthy donors. Subsequent screening of several AML cell lines revealed heterogeneous response to RITA, and confirmed an association of the specific phosphorylation with apoptosis. While decreased NCL phosphorylation at Threonines T76 and T84 could be attributed to RITA-induced cell cycle arrest, enhanced NPM phosphorylation at Threonine T199 was not accompanied by the cell cycle changes and it correlated with sensitivity to RITA. Simultaneously, inverse changes occurred at Serine S4 of the NPM. These new findings of RITA mechanism of action could establish the NPM pT199/pS4 ratio as a marker for suitability of RITA treatment of AML cells.
Collapse
Affiliation(s)
- Kateřina Wolfová
- Department of Proteomics, Institute of Hematology and Blood Transfusion, U Nemocnice 1, 12820 Prague 2, Czech Republic
| | - Petra Otevřelová
- Department of Proteomics, Institute of Hematology and Blood Transfusion, U Nemocnice 1, 12820 Prague 2, Czech Republic
| | - Aleš Holoubek
- Department of Proteomics, Institute of Hematology and Blood Transfusion, U Nemocnice 1, 12820 Prague 2, Czech Republic
| | - Barbora Brodská
- Department of Proteomics, Institute of Hematology and Blood Transfusion, U Nemocnice 1, 12820 Prague 2, Czech Republic.
| |
Collapse
|
42
|
Lee EJ, Choi JG, Han JH, Kim YW, Lim J, Chung HS. Single-Cell RNA Sequencing Reveals Immuno-Oncology Characteristics of Tumor-Infiltrating T Lymphocytes in Photodynamic Therapy-Treated Colorectal Cancer Mouse Model. Int J Mol Sci 2023; 24:13913. [PMID: 37762216 PMCID: PMC10531263 DOI: 10.3390/ijms241813913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Photodynamic therapy (PDT) has shown promise in reducing metastatic colorectal cancer (CRC); however, the underlying mechanisms remain unclear. Modulating tumor-infiltrating immune cells by PDT may be achieved, which requires the characterization of immune cell populations in the tumor microenvironment by single-cell RNA sequencing (scRNA-seq). Here, we determined the effect of Chlorin e6 (Ce6)-mediated PDT on tumor-infiltrating T cells using scRNA-seq analysis. We used a humanized programmed death-1/programmed death ligand 1 (PD-1/PD-L1) MC38 cell allograft mouse model, considering its potential as an immunogenic cancer model and in combination with PD-1/PD-L1 immune checkpoint blockade. PDT treatment significantly reduced tumor growth in mice containing hPD-1/PD-L1 MC38 tumors. scRNA-seq analysis revealed that the PDT group had increased levels of CD8+ activated T cells and CD8+ cytotoxic T cells, but decreased levels of exhausted CD8+ T cells. PDT treatment also enhanced the infiltration of CD8+ T cells into tumors and increased the production of key effector molecules, including granzyme B and perforin 1. These findings provide insight into immune-therapeutic modulation for CRC patients and highlight the potential of PDT in overcoming immune evasion and enhancing antitumor immunity.
Collapse
Affiliation(s)
- Eun-Ji Lee
- Korean Medicine Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 41062, Republic of Korea; (E.-J.L.); (J.-G.C.); (J.H.H.)
| | - Jang-Gi Choi
- Korean Medicine Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 41062, Republic of Korea; (E.-J.L.); (J.-G.C.); (J.H.H.)
| | - Jung Ho Han
- Korean Medicine Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 41062, Republic of Korea; (E.-J.L.); (J.-G.C.); (J.H.H.)
| | - Yong-Wan Kim
- Dongsung Cancer Center, Dongsung Biopharmaceutical, Daegu 41061, Republic of Korea; (Y.-W.K.); (J.L.)
| | - Junmo Lim
- Dongsung Cancer Center, Dongsung Biopharmaceutical, Daegu 41061, Republic of Korea; (Y.-W.K.); (J.L.)
| | - Hwan-Suck Chung
- Korean Medicine Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 41062, Republic of Korea; (E.-J.L.); (J.-G.C.); (J.H.H.)
- Korean Convergence Medical Science Major, University of Science and Technology (UST), KIOM Campus, Daegu 41062, Republic of Korea
| |
Collapse
|
43
|
Falini B. NPM1-mutated acute myeloid leukemia: New pathogenetic and therapeutic insights and open questions. Am J Hematol 2023; 98:1452-1464. [PMID: 37317978 DOI: 10.1002/ajh.26989] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 06/02/2023] [Indexed: 06/16/2023]
Abstract
The nucleophosmin (NPM1) gene encodes for a multifunctional chaperone protein that is localized in the nucleolus but continuously shuttles between the nucleus and cytoplasm. NPM1 mutations occur in about one-third of AML, are AML-specific, usually involve exon 12 and are frequently associated with FLT3-ITD, DNMT3A, TET2, and IDH1/2 mutations. Because of its unique molecular and clinico-pathological features, NPM1-mutated AML is regarded as a distinct leukemia entity in both the International Consensus Classification (ICC) and the 5th edition of the World Health Organization (WHO) classification of myeloid neoplasms. All NPM1 mutations generate leukemic mutants that are aberrantly exported in the cytoplasm of the leukemic cells and are relevant to the pathogenesis of the disease. Here, we focus on recently identified functions of the NPM1 mutant at chromatin level and its relevance in driving HOX/MEIS gene expression. We also discuss yet controversial issues of the ICC/WHO classifications, including the biological and clinical significance of therapy-related NPM1-mutated AML and the relevance of blasts percentage in defining NPM1-mutated AML. Finally, we address the impact of new targeted therapies in NPM1-mutated AML with focus on CAR T cells directed against NPM1/HLA neoepitopes, as well as XPO1 and menin inhibitors.
Collapse
Affiliation(s)
- Brunangelo Falini
- Institute of Hematology and Center for Hemato-Oncological Research (CREO), University of Perugia and Santa Maria della Misericordia Hospital, Perugia, Italy
| |
Collapse
|
44
|
Zeng T, Liao P, Zheng C, Gao H, Ye X, Zhou C, Zhou Y. The interaction between the lemon ribosomal protein ClRPS9-2 and citrus yellow vein clearing virus coat protein affects viral infection and gene silencing suppressor activity. MOLECULAR PLANT PATHOLOGY 2023; 24:1047-1062. [PMID: 37148475 PMCID: PMC10423326 DOI: 10.1111/mpp.13347] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 05/08/2023]
Abstract
Citrus yellow vein clearing virus (CYVCV) is an emerging virus that causes serious economic damage to the lemon industry worldwide. The coat protein (CP) of CYVCV is a strong RNA silencing suppressor and is associated with the severity of symptoms in citrus, yet the interaction between CP and host factors remains unknown. In this study, the 40S ribosomal subunit protein S9-2 (ClRPS9-2) was identified as a CP-binding partner using the yeast two-hybrid system from a lemon (cv. Eureka) cDNA library, and the interaction between CP and ClRPS9-2 was demonstrated by in vivo methods. The results suggest that the N-terminal 8-108 amino acid sequence of ClRPS9-2 is crucial for its interaction with CP and may be associated with the nuclear localization of ClRPS9-2. The accumulation and silencing suppressor activity of CP were reduced by transient expression of ClRPS9-2 in Nicotiana benthamiana. Reverse transcription-quantitative PCR analysis showed that the content of CYVCV in ClRPS9-2 transgenic Eureka lemon plants was approximately 50% of that in CYVCV-infected wild-type plants 1 month after inoculation, and mild yellowing and vein clearing symptoms were observed in the transgenic plants. These findings demonstrate that ClRPS9-2 plays a role in host defensive reactions, and the enhanced resistance of transgenic plants to CYVCV may be associated with the up-regulation of salicylic acid-related and R genes.
Collapse
Affiliation(s)
- Ting Zeng
- National Citrus Engineering Research CenterCitrus Research Institute, Southwest UniversityChongqingChina
| | - Ping Liao
- National Citrus Engineering Research CenterCitrus Research Institute, Southwest UniversityChongqingChina
| | - Cairong Zheng
- National Citrus Engineering Research CenterCitrus Research Institute, Southwest UniversityChongqingChina
| | - Haixing Gao
- National Citrus Engineering Research CenterCitrus Research Institute, Southwest UniversityChongqingChina
| | - Xiao Ye
- National Citrus Engineering Research CenterCitrus Research Institute, Southwest UniversityChongqingChina
| | - Changyong Zhou
- National Citrus Engineering Research CenterCitrus Research Institute, Southwest UniversityChongqingChina
| | - Yan Zhou
- National Citrus Engineering Research CenterCitrus Research Institute, Southwest UniversityChongqingChina
| |
Collapse
|
45
|
Ding X, Zhu XL, Xu DH, Li S, Yang Q, Feng X, Wei YG, Li H, Yang L, Zhang YJ, Deng XL, Liu KC, Shi SL. NPM promotes hepatotoxin-induced fibrosis by inhibiting ROS-induced apoptosis of hepatic stellate cells and upregulating lncMIAT-induced TGF-β2. Cell Death Dis 2023; 14:575. [PMID: 37648688 PMCID: PMC10469196 DOI: 10.1038/s41419-023-06043-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 07/25/2023] [Accepted: 08/04/2023] [Indexed: 09/01/2023]
Abstract
Liver fibrosis is caused by a variety of chronic liver injuries and has caused significant morbidity and mortality in the world with increasing tendency. Elucidation of the molecular mechanism of liver fibrosis is the basis for intervention of this pathological process and drug development. Nucleophosmin (NPM) is a widely expressed nucleolar phosphorylated protein, which is particularly important for cell proliferation, differentiation and survival. The biological role of NPM in liver fibrosis remains unknown. Here we show that NPM promotes liver fibrosis through multiple pathways. Our study found that NPM was up-regulated in cirrhosis tissues and activated in hepatic stellate cells (HSCs). NPM inhibition reduced liver fibrosis markers expression in HSCs and inhibited the HSCs proliferation and migration. In mice model, NPM knockdown in HSCs or application of specific NPM inhibitor can remarkably attenuate hepatic fibrosis. Mechanistic analysis showed that NPM promotes hepatic fibrosis by inhibiting HSCs apoptosis through Akt/ROS pathway and by upregulating TGF-β2 through Akt-induced lncMIAT. LncMIAT up-regulated TGF-β2 mRNA by competitively sponging miR-16-5p. In response to liver injury, hepatocytes, Kupffer cells and HSCs up-regulated NPM to increase TGF-β2 secretion to activate HSCs in a paracrine or autocrine manner, leading to increased liver fibrosis. Our study demonstrated that NPM regulated hepatotoxin-induced fibrosis through Akt/ROS-induced apoptosis of HSCs and via the Akt/lncMIAT-up-regulated TGF-β2. Inhibition of NPM or application of NPM inhibitor CIGB300 remarkably attenuated liver fibrosis. NPM serves a potential new drug target for liver fibrosis.
Collapse
Affiliation(s)
- Xue Ding
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China
| | - Xin-Le Zhu
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China
| | - Dong-Hui Xu
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
- Department of Hepatic Biliary Pancreatic Vascular Surgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Shuang Li
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Qiong Yang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Xian Feng
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Yong-Gui Wei
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Huan Li
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Ling Yang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Yu-Jun Zhang
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China
| | - Xiao-Ling Deng
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Kuan-Can Liu
- Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| | - Song-Lin Shi
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China.
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
46
|
Chen CH, Huang YM, Grillet L, Hsieh YC, Yang YW, Lo KY. Gallium maltolate shows synergism with cisplatin and activates nucleolar stress and ferroptosis in human breast carcinoma cells. Cell Oncol (Dordr) 2023; 46:1127-1142. [PMID: 37067747 DOI: 10.1007/s13402-023-00804-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2023] [Indexed: 04/18/2023] Open
Abstract
PURPOSE Breast cancer is the most common cancer in women. Triple-negative breast cancer (TNBC) is an aggressive disease with poor outcomes. TNBC lacks effective targeted treatments, and the development of drug resistance limits the effectiveness of chemotherapy. It is crucial to identify new drugs that can enhance the efficacy of traditional chemotherapy to reduce drug resistance and side effects. METHODS TNBC cell lines, MDA-MB-231 and Hs 578T, and a normal cell line, MCF-10 A, were included in this study. The cells were treated with gallium maltolate (GaM), and their transcriptome was analyzed. Ferroptosis and nucleolar stress markers were detected by qPCR, western blotting, fluorescence microscopy, and flow cytometry. The impairment of ribosome synthesis was evaluated by northern blotting and sucrose gradients. RESULTS GaM triggered cell death via apoptosis and ferroptosis. In addition, GaM impaired translation and activated nucleolar stress. Cisplatin (DDP) is a chemotherapeutic agent for advanced breast cancer. While single treatment with GaM or DDP at low concentrations did not impact cell growth, co-administration enhanced cell death in TNBC but not in normal breast cells. The enhancement of ferroptosis and nucleolar stress could be observed in TNBC cell lines after co-treatment. CONCLUSIONS These results suggest that GaM synergizes with cisplatin via activation of nucleolar stress and ferroptosis in human breast carcinoma cells. GaM is marginally toxic to normal cells but impairs the growth of TNBC cell lines. Thus, GaM has the potential to be used as a therapeutic agent against TNBC.
Collapse
Affiliation(s)
- Chieh-Hsin Chen
- Department of Agricultural Chemistry, National Taiwan University, 1 Sec. 4, Roosevelt Road, Taipei, 6836, 10617, Taiwan
| | - Yi-Ming Huang
- Department of Agricultural Chemistry, National Taiwan University, 1 Sec. 4, Roosevelt Road, Taipei, 6836, 10617, Taiwan
| | - Louis Grillet
- Department of Agricultural Chemistry, National Taiwan University, 1 Sec. 4, Roosevelt Road, Taipei, 6836, 10617, Taiwan
| | - Yu-Chen Hsieh
- Department of Agricultural Chemistry, National Taiwan University, 1 Sec. 4, Roosevelt Road, Taipei, 6836, 10617, Taiwan
| | - Ya-Wen Yang
- Department of Surgery, National Taiwan University Hospital, No.7, Chung Shan S. Rd., Zhongzheng Dist, Taipei City, 100225, Taiwan.
| | - Kai-Yin Lo
- Department of Agricultural Chemistry, National Taiwan University, 1 Sec. 4, Roosevelt Road, Taipei, 6836, 10617, Taiwan.
| |
Collapse
|
47
|
Park SW, Park IB, Kang SJ, Bae J, Chun T. Interaction between host cell proteins and open reading frames of porcine circovirus type 2. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2023; 65:698-719. [PMID: 37970506 PMCID: PMC10640953 DOI: 10.5187/jast.2023.e67] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/28/2023] [Accepted: 07/09/2023] [Indexed: 11/17/2023]
Abstract
Postweaning multisystemic wasting syndrome (PMWS) is caused by a systemic inflammation after porcine circovirus type 2 (PCV2) infection. It was one of the most economically important pathogens affecting pig production worldwide before PCV2 vaccine was first introduced in 2006. After the development of a vaccine against PCV2a type, pig farms gradually restored enormous economic losses from PMWS. However, vaccine against PCV2a type could not be fully effective against several different PCV2 genotypes (PCV2b - PCV2h). In addition, PCV2a vaccine itself could generate antigenic drift of PCV2 capsid. Therefore, PCV2 infection still threats pig industry worldwide. PCV2 infection was initially found in local tissues including reproductive, respiratory, and digestive tracks. However, PCV2 infection often leads to a systemic inflammation which can cause severe immunosuppression by depleting peripheral lymphocytes in secondary lymphoid tissues. Subsequently, a secondary infection with other microorganisms can cause PMWS. Eleven putative open reading frames (ORFs) have been predicted to encode PCV2 genome. Among them, gene products of six ORFs from ORF1 to ORF6 have been identified and characterized to estimate its functional role during PCV2 infection. Acquiring knowledge about the specific interaction between each PCV2 ORF protein and host protein might be a key to develop preventive or therapeutic tools to control PCV2 infection. In this article, we reviewed current understanding of how each ORF of PCV2 manipulates host cell signaling related to immune suppression caused by PCV2.
Collapse
Affiliation(s)
- Si-Won Park
- Department of Biotechnology, School of
Life Sciences and Biotechnology, Korea University, Seoul
02841, Korea
| | - In-Byung Park
- Department of Biotechnology, School of
Life Sciences and Biotechnology, Korea University, Seoul
02841, Korea
| | - Seok-Jin Kang
- Department of Biotechnology, School of
Life Sciences and Biotechnology, Korea University, Seoul
02841, Korea
| | - Joonbeom Bae
- Department of Biotechnology, School of
Life Sciences and Biotechnology, Korea University, Seoul
02841, Korea
| | - Taehoon Chun
- Department of Biotechnology, School of
Life Sciences and Biotechnology, Korea University, Seoul
02841, Korea
| |
Collapse
|
48
|
Mill CP, Fiskus W, Das K, Davis JA, Birdwell CE, Kadia TM, DiNardo CD, Daver N, Takahashi K, Sasaki K, McGeehan GM, Ruan X, Su X, Loghavi S, Kantarjian H, Bhalla KN. Causal linkage of presence of mutant NPM1 to efficacy of novel therapeutic agents against AML cells with mutant NPM1. Leukemia 2023; 37:1336-1348. [PMID: 36977823 PMCID: PMC10244173 DOI: 10.1038/s41375-023-01882-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023]
Abstract
In AML with NPM1 mutation causing cytoplasmic dislocation of NPM1, treatments with Menin inhibitor (MI) and standard AML chemotherapy yield complete remissions. However, the causal and mechanistic linkage of mtNPM1 to the efficacy of these agents has not been definitively established. Utilizing CRISPR-Cas9 editing to knockout (KO) or knock-in a copy of mtNPM1 in AML cells, present studies demonstrate that KO of mtNPM1 from AML cells abrogates sensitivity to MI, selinexor (exportin-1 inhibitor), and cytarabine. Conversely, the knock-in of a copy of mtNPM1 markedly sensitized AML cells to treatment with MI or cytarabine. Following AML therapy, most elderly patients with AML with mtNPM1 and co-mutations in FLT3 suffer AML relapse with poor outcomes, creating a need for novel effective therapies. Utilizing the RNA-Seq signature of CRISPR-edited AML cells with mtNPM1 KO, we interrogated the LINCS1000-CMap data set and found several pan-HDAC inhibitors and a WEE1 tyrosine kinase inhibitor among the top expression mimickers (EMs). Additionally, treatment with adavosertib (WEE1 inhibitor) or panobinostat (pan-HDAC inhibitor) exhibited synergistic in vitro lethal activity with MI against AML cells with mtNPM1. Treatment with adavosertib or panobinostat also reduced AML burden and improved survival in AML xenograft models sensitive or resistant to MI.
Collapse
Affiliation(s)
- Christopher P Mill
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Warren Fiskus
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kaberi Das
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - John A Davis
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | | | - Tapan M Kadia
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Courtney D DiNardo
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Naval Daver
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Koichi Takahashi
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Koji Sasaki
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | | | - Xinjia Ruan
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xiaoping Su
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sanam Loghavi
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hagop Kantarjian
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kapil N Bhalla
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
49
|
Sakthivel D, Brown-Suedel AN, Keane F, Huang S, Sherry KM, Charendoff CI, Dunne KP, Robichaux DJ, Le B, Shin CS, Carisey AF, Flanagan JM, Bouchier-Hayes L. Caspase-2 is essential for proliferation and self-renewal of nucleophosmin-mutated acute myeloid leukemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.29.542723. [PMID: 37398413 PMCID: PMC10312440 DOI: 10.1101/2023.05.29.542723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Mutation in nucleophosmin (NPM1) causes relocalization of this normally nucleolar protein to the cytoplasm ( NPM1c+ ). Despite NPM1 mutation being the most common driver mutation in cytogenetically normal adult acute myeloid leukemia (AML), the mechanisms of NPM1c+-induced leukemogenesis remain unclear. Caspase-2 is a pro-apoptotic protein activated by NPM1 in the nucleolus. Here, we show that caspase-2 is also activated by NPM1c+ in the cytoplasm, and DNA damage-induced apoptosis is caspase-2-dependent in NPM1c+ AML but not in NPM1wt cells. Strikingly, in NPM1c+ cells, loss of caspase-2 results in profound cell cycle arrest, differentiation, and down-regulation of stem cell pathways that regulate pluripotency including impairment in the AKT/mTORC1 and Wnt signaling pathways. In contrast, there were minimal differences in proliferation, differentiation, or the transcriptional profile of NPM1wt cells with and without caspase-2. Together, these results show that caspase-2 is essential for proliferation and self-renewal of AML cells that have mutated NPM1. This study demonstrates that caspase-2 is a major effector of NPM1c+ function and may even be a druggable target to treat NPM1c+ AML and prevent relapse.
Collapse
|
50
|
Yang G, Li H, Dong Z, Deng K, Lu Y. Nucleophosmin 1 associating with engulfment and cell motility protein 1 regulates hepatocellular carcinoma cell chemotaxis and metastasis. Open Med (Wars) 2023; 18:20230708. [PMID: 37251542 PMCID: PMC10224614 DOI: 10.1515/med-2023-0708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 03/24/2023] [Accepted: 04/17/2023] [Indexed: 05/31/2023] Open
Abstract
The chemokine, C-X-C motif chemokine ligand 12 (CXCL12) and its G-protein-coupled receptor (GPCR) and C-X-C chemokine receptor type 4 (CXCR4), are closely associated with promoting hepatocellular carcinoma (HCC) chemotaxis and metastasis. The binding of CXCL12 and CXCR4 depends on the heterotrimeric Gi proteins to regulate actin polymerisation and mobility in HCC. Although the role of GPCR/Gi signalling in carcinogenesis migration has been intensively studied, the detailed mechanism remains largely unknown. In this study, a small interfering RNA technique was used to knock down the Nucleophosmin 1 (NPM1) gene expression. Through the chemotaxis and invasion assays, wound healing, proliferation, filamentous-actin, immunofluorescence, immunohistochemical assays, and co-immunoprecipitation assays, we investigated the specific biological role and underlying mechanisms of the NPM1 in HCC. Additionally, dimethyl fumarate (DMF), a fumaric acid ester, was used to inhibit the HCC cell chemokines and metastasis by regulating ELMO1 and NPM1. Therefore, this study reported that NPM1 gene expression was upregulated in the HCC tissues and cell lines. The NPM1 knockdown significantly inhibited the proliferation, migration, and chemotaxis of the HepG2 cells in vitro. Further mechanistic studies suggested that the NPM1 interacts with ELMO1 and the CXCL12/CXCR4 pathway activates NPM1-dependent regulation of the ELMO1 localisation. Furthermore, the DMF significantly inhibited tumour metastasis induced by the NPM1/ELMO1 signalling pathway, as observed in in vitro cell functional experiments. These data suggested that as a potentially novel therapeutic approach, the simultaneous targeting of NPM1 and ELMO1 could effectively be used to treat HCC.
Collapse
Affiliation(s)
- Gangqi Yang
- Department of Infectious Diseases, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, China
- General Surgery Department and Neurology Department, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Beijing100053, China
- Comprehensive Liver Cancer Center, 5th Medical Center of the PLA General Hospital, Beijing100039, China
- Guangdong Key Laboratory of Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Hongyan Li
- General Surgery Department and Neurology Department, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Beijing100053, China
| | - Zheng Dong
- Comprehensive Liver Cancer Center, 5th Medical Center of the PLA General Hospital, Beijing100039, China
| | - Kai Deng
- The First Affiliated Hospital of Chongqing Medical College, Chongqing400016, China
| | - Yinying Lu
- Department of Infectious Diseases, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, China
- Comprehensive Liver Cancer Center, 5th Medical Center of the PLA General Hospital, Beijing100039, China
- Guangdong Key Laboratory of Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong 518055, China
| |
Collapse
|