1
|
Deng H, Xie K, Hu L, Liu X, Li Q, Xie D, Xiang F, Liu W, Zheng W, Xiao S, Zheng J, Tan X. Polyamine Derived Photosensitizer: A Novel Approach for Photodynamic Therapy of Cancer. Molecules 2024; 29:4277. [PMID: 39275124 PMCID: PMC11397399 DOI: 10.3390/molecules29174277] [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: 05/10/2024] [Revised: 05/28/2024] [Accepted: 08/30/2024] [Indexed: 09/16/2024] Open
Abstract
Polyamines play a pivotal role in cancer cell proliferation. The excessive polyamine requirement of these malignancies is satisfied through heightened biosynthesis and augmented extracellular uptake via the polyamine transport system (PTS) present on the cell membrane. Meanwhile, photodynamic therapy (PDT) emerges as an effective anti-cancer treatment devoid of drug resistance. Recognizing these intricacies, our study devised a novel polyamine-derived photosensitizer (PS) for targeted photodynamic treatment, focusing predominantly on pancreatic cancer cells. We synthesized and evaluated novel spermine-derived fluorescent probes (N2) and PS (N3), exhibiting selectivity towards pancreatic cancer cells via PTS. N3 showed minimal dark toxicity but significant phototoxicity upon irradiation, effectively causing cell death in vitro. A significant reduction in tumor volume was observed post-treatment with no pronounced dark toxicity using the pancreatic cancer CDX mouse model, affirming the therapeutic potential of N3. Overall, our findings introduce a promising new strategy for cancer treatment, highlighting the potential of polyamine-derived PSs in PDT.
Collapse
Affiliation(s)
- Hao Deng
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- The First College of Clinical Medical Science, China Three Gorges University, Yichang 443003, China
| | - Ke Xie
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- The First College of Clinical Medical Science, China Three Gorges University, Yichang 443003, China
| | - Liling Hu
- The First College of Clinical Medical Science, China Three Gorges University, Yichang 443003, China
| | - Xiaowen Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Qingyun Li
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Donghui Xie
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Fengyi Xiang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Wei Liu
- The First College of Clinical Medical Science, China Three Gorges University, Yichang 443003, China
| | - Weihong Zheng
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Shuzhang Xiao
- College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Jun Zheng
- The First College of Clinical Medical Science, China Three Gorges University, Yichang 443003, China
| | - Xiao Tan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- The First College of Clinical Medical Science, China Three Gorges University, Yichang 443003, China
| |
Collapse
|
2
|
Vincelette ND, Yu X, Kuykendall AT, Moon J, Su S, Cheng CH, Sammut R, Razabdouski TN, Nguyen HV, Eksioglu EA, Chan O, Al Ali N, Patel PC, Lee DH, Nakanishi S, Ferreira RB, Hyjek E, Mo Q, Cory S, Lawrence HR, Zhang L, Murphy DJ, Komrokji RS, Lee D, Kaufmann SH, Cleveland JL, Yun S. Trisomy 8 Defines a Distinct Subtype of Myeloproliferative Neoplasms Driven by the MYC-Alarmin Axis. Blood Cancer Discov 2024; 5:276-297. [PMID: 38713018 PMCID: PMC11215389 DOI: 10.1158/2643-3230.bcd-23-0210] [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: 10/09/2023] [Revised: 03/16/2024] [Accepted: 05/06/2024] [Indexed: 05/08/2024] Open
Abstract
Despite advances in understanding the genetic abnormalities in myeloproliferative neoplasms (MPN) and the development of JAK2 inhibitors, there is an urgent need to devise new treatment strategies, particularly for patients with triple-negative (TN) myelofibrosis (MF) who lack mutations in the JAK2 kinase pathway and have very poor clinical outcomes. Here we report that MYC copy number gain and increased MYC expression frequently occur in TN-MF and that MYC-directed activation of S100A9, an alarmin protein that plays pivotal roles in inflammation and innate immunity, is necessary and sufficient to drive development and progression of MF. Notably, the MYC-S100A9 circuit provokes a complex network of inflammatory signaling that involves numerous hematopoietic cell types in the bone marrow microenvironment. Accordingly, genetic ablation of S100A9 or treatment with small molecules targeting the MYC-S100A9 pathway effectively ameliorates MF phenotypes, highlighting the MYC-alarmin axis as a novel therapeutic vulnerability for this subgroup of MPNs. Significance: This study establishes that MYC expression is increased in TN-MPNs via trisomy 8, that a MYC-S100A9 circuit manifest in these cases is sufficient to provoke myelofibrosis and inflammation in diverse hematopoietic cell types in the BM niche, and that the MYC-S100A9 circuit is targetable in TN-MPNs.
Collapse
Affiliation(s)
- Nicole D. Vincelette
- Department of Malignant Hematology, Moffitt Cancer Center & Research Institute, Tampa, Florida.
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center & Research Institute, Tampa, Florida.
| | - Andrew T. Kuykendall
- Department of Malignant Hematology, Moffitt Cancer Center & Research Institute, Tampa, Florida.
| | - Jungwon Moon
- Department of Malignant Hematology, Moffitt Cancer Center & Research Institute, Tampa, Florida.
| | - Siyuan Su
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois.
| | - Chia-Ho Cheng
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center & Research Institute, Tampa, Florida.
| | - Rinzine Sammut
- Department of Malignant Hematology, Moffitt Cancer Center & Research Institute, Tampa, Florida.
- Département d’Hématologie Clinique, Centre Hospitalier Universitaire de Nice, Nice, France.
| | - Tiffany N. Razabdouski
- Department of Malignant Hematology, Moffitt Cancer Center & Research Institute, Tampa, Florida.
| | - Hai V. Nguyen
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.
| | - Erika A. Eksioglu
- Department of Immunology, Moffitt Cancer Center & Research Institute, Tampa, Florida.
| | - Onyee Chan
- Department of Malignant Hematology, Moffitt Cancer Center & Research Institute, Tampa, Florida.
| | - Najla Al Ali
- Department of Malignant Hematology, Moffitt Cancer Center & Research Institute, Tampa, Florida.
| | - Parth C. Patel
- Department of Malignant Hematology, Moffitt Cancer Center & Research Institute, Tampa, Florida.
- Department of Internal Medicine, University of South Florida, Tampa, Florida.
| | - Dae H. Lee
- Division of Cardiovascular Science, Department of Internal Medicine, University of South Florida, Tampa, Florida
| | - Shima Nakanishi
- Department of Tumor Microenvironment & Metastasis, Moffitt Cancer Center & Research Institute, Tampa, Florida.
| | - Renan B. Ferreira
- Department of Drug Discovery, Moffitt Cancer Center & Research Institute, Tampa, Florida.
| | - Elizabeth Hyjek
- Department of Pathology and Laboratory Medicine, Moffitt Cancer Center & Research Institute, Tampa, Florida.
| | - Qianxing Mo
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center & Research Institute, Tampa, Florida.
| | - Suzanne Cory
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.
| | - Harshani R. Lawrence
- Department of Drug Discovery, Moffitt Cancer Center & Research Institute, Tampa, Florida.
| | - Ling Zhang
- Department of Pathology and Laboratory Medicine, Moffitt Cancer Center & Research Institute, Tampa, Florida.
| | - Daniel J. Murphy
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom.
- Cancer Research UK Scotland Institute, Glasgow, United Kingdom.
| | - Rami S. Komrokji
- Department of Malignant Hematology, Moffitt Cancer Center & Research Institute, Tampa, Florida.
| | - Daesung Lee
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois.
| | - Scott H. Kaufmann
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota.
- Department of Oncology, Mayo Clinic, Rochester, Minnesota.
| | - John L. Cleveland
- Department of Tumor Microenvironment & Metastasis, Moffitt Cancer Center & Research Institute, Tampa, Florida.
| | - Seongseok Yun
- Department of Malignant Hematology, Moffitt Cancer Center & Research Institute, Tampa, Florida.
| |
Collapse
|
3
|
Holbert CE, Casero RA, Stewart TM. Polyamines: the pivotal amines in influencing the tumor microenvironment. Discov Oncol 2024; 15:173. [PMID: 38761252 PMCID: PMC11102423 DOI: 10.1007/s12672-024-01034-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 05/11/2024] [Indexed: 05/20/2024] Open
Abstract
Cellular proliferation, function and survival is reliant upon maintaining appropriate intracellular polyamine levels. Due to increased metabolic needs, cancer cells elevate their polyamine pools through coordinated metabolism and uptake. High levels of polyamines have been linked to more immunosuppressive tumor microenvironments (TME) as polyamines support the growth and function of many immunosuppressive cell types such as MDSCs, macrophages and regulatory T-cells. As cancer cells and other pro-tumorigenic cell types are highly dependent on polyamines for survival, pharmacological modulation of polyamine metabolism is a promising cancer therapeutic strategy. This review covers the roles of polyamines in various cell types of the TME including both immune and stromal cells, as well as how competition for nutrients, namely polyamine precursors, influences the cellular landscape of the TME. It also details the use of polyamines as biomarkers and the ways in which polyamine depletion can increase the immunogenicity of the TME and reprogram tumors to become more responsive to immunotherapy.
Collapse
Affiliation(s)
- Cassandra E Holbert
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Robert A Casero
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Tracy Murray Stewart
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
4
|
Xuan M, Gu X, Li J, Huang D, Xue C, He Y. Polyamines: their significance for maintaining health and contributing to diseases. Cell Commun Signal 2023; 21:348. [PMID: 38049863 PMCID: PMC10694995 DOI: 10.1186/s12964-023-01373-0] [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: 08/26/2023] [Accepted: 10/29/2023] [Indexed: 12/06/2023] Open
Abstract
Polyamines are essential for the growth and proliferation of mammalian cells and are intimately involved in biological mechanisms such as DNA replication, RNA transcription, protein synthesis, and post-translational modification. These mechanisms regulate cellular proliferation, differentiation, programmed cell death, and the formation of tumors. Several studies have confirmed the positive effect of polyamines on the maintenance of health, while others have demonstrated that their activity may promote the occurrence and progression of diseases. This review examines a variety of topics, such as polyamine source and metabolism, including metabolism, transport, and the potential impact of polyamines on health and disease. In addition, a brief summary of the effects of oncogenes and signaling pathways on tumor polyamine metabolism is provided. Video Abstract.
Collapse
Affiliation(s)
- Mengjuan Xuan
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Xinyu Gu
- Department of Oncology, College of Clinical Medicine, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Juan Li
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Di Huang
- Department of Child Health Care, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Chen Xue
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Yuting He
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
5
|
Shen X, Dong P, Kong J, Sun N, Wang F, Sang L, Xu Y, Zhang M, Chen X, Guo R, Wang S, Lin Q, Jiang Z, Xu S, Zhang C, Bian Z, Wang W, Guo R. Targeted single-cell RNA sequencing analysis reveals metabolic reprogramming and the ferroptosis-resistant state in hematologic malignancies. Cell Biochem Funct 2023; 41:1343-1356. [PMID: 37823726 DOI: 10.1002/cbf.3869] [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: 07/11/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/13/2023]
Abstract
Hematologic malignancies are the most common hematopoietic diseases and a major public health concern. However, the mechanisms underlying myeloid tumors remain unknown owing to the intricate interplay between mutations and diverse clonal evolution patterns, as evidenced by the analysis of bulk cell-derived omics data. Several single-cell omics techniques have been used to characterize the hierarchies and altered immune microenvironments of hematologic malignancies. The comprehensive single-cell atlas of hematologic malignancies provides novel opportunities for personalized combinatorial targeted treatments, avoiding unwanted chemo-toxicity. In the present study, we performed transcriptome sequencing by combining single-cell RNA sequencing (scRNA-seq) with a targeted oncogenic gene panel for acute myeloid leukemia, overcoming the limitations of scRNA-seq in detecting oncogenic mutations. The distribution of oncogenic IDH1, IDH2, and KRAS mutations in each cell type was identified in the bone marrow (BM) samples of each patient. Our findings suggest that ferroptosis and metabolic reprogramming are involved in the tumorigenesis and chemotherapy resistance of oncogenic mutation-carrying cells. Biological progression via IDH1, IDH2, and KRAS mutations arrests hematopoietic maturation. Our study findings provide a rationale for using primary BM cells for personalized treatment in clinical settings.
Collapse
Affiliation(s)
- Xiaohui Shen
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peiyuan Dong
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jingjing Kong
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Nannan Sun
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fang Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lina Sang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yan Xu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengmeng Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoli Chen
- Jiangxi Health Commission Key Laboratory of Leukemia, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi, China
| | - Rong Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuya Wang
- Department of Blood Transfusion, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Quande Lin
- Department of Hematology, The Afliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Zhongxing Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shan Xu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Congli Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhilei Bian
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Weimin Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Rongqun Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
6
|
Bailleux C, Chardin D, Gal J, Guigonis JM, Lindenthal S, Graslin F, Arnould L, Cagnard A, Ferrero JM, Humbert O, Pourcher T. Metabolomic Signatures of Scarff-Bloom-Richardson (SBR) Grade in Non-Metastatic Breast Cancer. Cancers (Basel) 2023; 15:cancers15071941. [PMID: 37046602 PMCID: PMC10093598 DOI: 10.3390/cancers15071941] [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: 02/06/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
PURPOSE Identification of metabolomic biomarkers of high SBR grade in non-metastatic breast cancer. METHODS This retrospective bicentric metabolomic analysis included a training set (n = 51) and a validation set (n = 49) of breast cancer tumors, all classified as high-grade (grade III) or low-grade (grade I-II). Metabolomes of tissue samples were studied by liquid chromatography coupled with mass spectrometry. RESULTS A molecular signature of the top 12 metabolites was identified from a database of 602 frequently predicted metabolites. Partial least squares discriminant analyses showed that accuracies were 0.81 and 0.82, the R2 scores were 0.57 and 0.55, and the Q2 scores were 0.44431 and 0.40147 for the training set and validation set, respectively; areas under the curve for the Receiver Operating Characteristic Curve were 0.882 and 0.886. The most relevant metabolite was diacetylspermine. Metabolite set enrichment analyses and metabolic pathway analyses highlighted the tryptophan metabolism pathway, but the concentration of individual metabolites varied between tumor samples. CONCLUSIONS This study indicates that high-grade invasive tumors are related to diacetylspermine and tryptophan metabolism, both involved in the inhibition of the immune response. Targeting these pathways could restore anti-tumor immunity and have a synergistic effect with immunotherapy. Recent studies could not demonstrate the effectiveness of this strategy, but the use of theragnostic metabolomic signatures should allow better selection of patients.
Collapse
Affiliation(s)
- Caroline Bailleux
- Laboratory Transporter in Imaging and Radiotherapy in Oncology (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Fréderic Joliot, Commissariat à l'Energie Atomique et aux Énergies Alternatives (CEA), Université Côte d'Azur (UCA), 06100 Nice, France
- Medical Oncology Department, Centre Antoine Lacassagne, University Côte d'Azur, 06189 Nice, France
| | - David Chardin
- Laboratory Transporter in Imaging and Radiotherapy in Oncology (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Fréderic Joliot, Commissariat à l'Energie Atomique et aux Énergies Alternatives (CEA), Université Côte d'Azur (UCA), 06100 Nice, France
- Department of Nuclear Medicine, Antoine Lacassagne Centre, 06189 Nice, France
| | - Jocelyn Gal
- Department of Epidemiology and Biostatistics, Antoine Lacassagne Centre, University of Côte d'Azur, 06189 Nice, France
| | - Jean-Marie Guigonis
- Laboratory Transporter in Imaging and Radiotherapy in Oncology (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Fréderic Joliot, Commissariat à l'Energie Atomique et aux Énergies Alternatives (CEA), Université Côte d'Azur (UCA), 06100 Nice, France
| | - Sabine Lindenthal
- Laboratory Transporter in Imaging and Radiotherapy in Oncology (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Fréderic Joliot, Commissariat à l'Energie Atomique et aux Énergies Alternatives (CEA), Université Côte d'Azur (UCA), 06100 Nice, France
| | - Fanny Graslin
- Laboratory Transporter in Imaging and Radiotherapy in Oncology (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Fréderic Joliot, Commissariat à l'Energie Atomique et aux Énergies Alternatives (CEA), Université Côte d'Azur (UCA), 06100 Nice, France
- Department of Nuclear Medicine, Antoine Lacassagne Centre, 06189 Nice, France
| | - Laurent Arnould
- Department of Tumour Biology and Pathology, Georges-François Leclerc Centre, 21079 Dijon, France
- Cenre de Ressources Biologiques (CRB) Ferdinand Cabanne, 21000 Dijon, France
| | - Alexandre Cagnard
- Laboratory Transporter in Imaging and Radiotherapy in Oncology (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Fréderic Joliot, Commissariat à l'Energie Atomique et aux Énergies Alternatives (CEA), Université Côte d'Azur (UCA), 06100 Nice, France
| | - Jean-Marc Ferrero
- Medical Oncology Department, Centre Antoine Lacassagne, University Côte d'Azur, 06189 Nice, France
| | - Olivier Humbert
- Laboratory Transporter in Imaging and Radiotherapy in Oncology (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Fréderic Joliot, Commissariat à l'Energie Atomique et aux Énergies Alternatives (CEA), Université Côte d'Azur (UCA), 06100 Nice, France
- Department of Nuclear Medicine, Antoine Lacassagne Centre, 06189 Nice, France
| | - Thierry Pourcher
- Laboratory Transporter in Imaging and Radiotherapy in Oncology (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Fréderic Joliot, Commissariat à l'Energie Atomique et aux Énergies Alternatives (CEA), Université Côte d'Azur (UCA), 06100 Nice, France
| |
Collapse
|
7
|
Uslu Bıçak İ, Tokcan B, Yavuz AS, Sözer Tokdemir S. Circulating CD133+/–CD34– Have Increased c- MYC Expression in Myeloproliferative Neoplasms. Turk J Haematol 2023; 40:28-36. [PMID: 36458557 PMCID: PMC9979741 DOI: 10.4274/tjh.galenos.2022.2022.0343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Objective Myeloproliferative neoplasms (MPNs) are hematopoietic stem cell (HSC)-originated diseases with clonal myeloproliferation. The constitutive activation of the JAK/STAT pathway is frequently detected in patients with Philadelphia chromosome-negative (Ph–) MPNs with an acquired JAK2V617F mutation. The c-MYC proto-oncogene is associated with malignant growth and cellular transformation, and JAK2V617F was previously shown to induce constitutive expression of c-MYC. This study examines the expressional profile of c-MYC in Ph– MPNs with JAK2V617F and highlights its hierarchical level of activation in circulating hematopoietic stem/progenitor cell (HSPC) subgroups. Materials and Methods Mononuclear cells (MNCs) of Ph– MPNs were fluorochrome-labeled in situ with wild-type (wt) JAK2 or JAK2V617F mRNA gold nanoparticle technology and sorted simultaneously. Isolated populations of JAK2wt or JAK2V617F were evaluated for their c-MYC expressions. The MNCs of 14 Ph– MPNs were further isolated for the study of HSPC subgroups regarding their CD34 and CD133 expressions, evaluated for the presence of JAK2V617F, and compared to cord blood (CB) counterparts for the expression of c-MYC. Results The mRNA-labeled gold nanoparticle-treated MNCs were determined to have the highest ratio of c-MYC relative fold-change expression in the biallelic JAK2V617F compartment compared to JAK2wt. The relative c-MYC expression in MNCs of MPNs was significantly increased compared to CB (p=0.01). The circulating HSPCs of CD133+/–CD34− MPNs had statistically significantly elevated c-MYC expression compared to CB. Conclusion This is the first study of circulating CD133+/–CD34− cells in Ph– MPNs and it has revealed elevated c-MYC expression levels in HSCs/endothelial progenitor cells (HSCs/EPCs) and EPCs. Furthermore, the steady increase in the expression of c-MYC within MNCs carrying no mutations and monoallelic or biallelic JAK2V617F transcripts was notable. The presence of JAK2V617F with respect to c-MYC expression in the circulating HSCs/EPCs and EPCs of MPNs might provide some evidence for the initiation of JAK2V617F and propagation of disease. Further studies are needed to clarify the implications of increased c-MYC expression in such populations.
Collapse
Affiliation(s)
- İldeniz Uslu Bıçak
- İstanbul University Aziz Sancar Institute of Experimental Medicine, Department of Genetics, İstanbul, Türkiye,İstanbul University, Institute of Health Sciences, İstanbul, Türkiye
| | - Berkay Tokcan
- İstanbul University Aziz Sancar Institute of Experimental Medicine, Department of Genetics, İstanbul, Türkiye,İstanbul University, Institute of Health Sciences, İstanbul, Türkiye
| | - Akif Selim Yavuz
- İstanbul University Faculty of Medicine, Department of Internal Medicine, Division of Hematology, İstanbul, Türkiye
| | - Selçuk Sözer Tokdemir
- İstanbul University Aziz Sancar Institute of Experimental Medicine, Department of Genetics, İstanbul, Türkiye,* Address for Correspondence: İstanbul University Aziz Sancar Institute of Experimental Medicine, Department of Genetics, İstanbul, Türkiye E-mail:
| |
Collapse
|
8
|
Calabresi L, Balliu M, Bartalucci N. Immunoblotting-assisted assessment of JAK/STAT and PI3K/Akt/mTOR signaling in myeloproliferative neoplasms CD34+ stem cells. Methods Cell Biol 2022; 171:81-109. [DOI: 10.1016/bs.mcb.2022.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
9
|
Zhu Y, Piao C, Zhang Z, Jiang Y, Kong C. The potential role of c-MYC and polyamine metabolism in multiple drug resistance in bladder cancer investigated by metabonomics. Genomics 2021; 114:125-137. [PMID: 34843906 DOI: 10.1016/j.ygeno.2021.11.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 11/15/2021] [Accepted: 11/23/2021] [Indexed: 12/11/2022]
Abstract
Bladder cancer has a high incidence worldwide accompanies by high recurrent rate after treatment. The emergence of primary or acquired chemotherapy resistance leads to poor efficacy in many cases. To explore the underlying mechanisms of drug resistance, we firstly established a drug-resistant cell model T24/THP by repeated exposure of T24 cells to pirarubicin (THP) whose concentration increases gradually. Non-targeted metabolomics was performed to identify metabolic changes and key metabolism pathways variance in T24/THP cells. Pathway enrichment analysis demonstrated that the arginine and proline metabolic pathway was the most significantly changed pathway, where two representative members of polyamine, putrescine and spermidine were remarkably down regulated in T24/THP. Subsequent experiments further confirmed that ornithine decarboxylase (ODC1) and spermidine synthase (SRM), the key enzymes involved in the synthesis of these compounds, also showed a stable low expression in T24/THP. However, knocking down of ODC1 and SRM sensitized cells to chemotherapy treatment while overexpression of these two enzymes enhances chemotherapy resistance. This leaded to the point that ODC1 and SRM themselves are more likely to promote the drug resistance, which appears to contradict their low expression in T24/THP. We hypothesize that their diminished levels were due to the declined activity of genes upstream. According to this line of thought, we found that c-MYC was also down-regulated in T24/THP and its content could be significantly affected by drug administration. In addition, c-MYC could not only regulate the expression levels of ODC1 and SRM but also influence drug resistance in T24/THP. In conclusion, alterations in gene expression of ODC1 and SRM in drug resistance cell line is probably mediated by some upstream regulators rather than antineoplastic agents alone. Exploration of upstream signals and research on detailed regulatory mechanism, thereby understanding the actual role of c-MYC and polyamine in response to chemotherapy, can become a potential field direction to overcome drug resistance in bladder cancer.
Collapse
Affiliation(s)
- Yiming Zhu
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, PR China
| | - Chiyuan Piao
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, PR China
| | - Zhe Zhang
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, PR China
| | - Yuanjun Jiang
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, PR China..
| | - Chuize Kong
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, PR China..
| |
Collapse
|
10
|
Tipgomut C, Khuhapinant A, Wilson MC, Poldee S, Heesom KJ, Metheetrairut C, Sripichai O, Mitrpant C, Frayne J, Trakarnsanga K. MTAP-related increased erythroblast proliferation as a mechanism of polycythaemia vera. Sci Rep 2021; 11:22483. [PMID: 34795367 PMCID: PMC8602418 DOI: 10.1038/s41598-021-01877-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/08/2021] [Indexed: 12/04/2022] Open
Abstract
Polycythaemia vera (PV) is a haematological disorder caused by an overproduction of erythroid cells. To date, the molecular mechanisms involved in the disease pathogenesis are still ambiguous. This study aims to identify aberrantly expressed proteins in erythroblasts of PV patients by utilizing mass spectrometry-based proteomic analysis. Haematopoietic stem cells (HSCs) were isolated from newly-diagnosed PV patients, PV patients who have received cytoreductive therapy, and healthy subjects. In vitro erythroblast expansion confirmed that the isolated HSCs recapitulated the disease phenotype as the number of erythroblasts from newly-diagnosed PV patients was significantly higher than those from the other groups. Proteomic comparison revealed 17 proteins that were differentially expressed in the erythroblasts from the newly-diagnosed PV patients compared to those from healthy subjects, but which were restored to normal levels in the patients who had received cytoreductive therapy. One of these proteins was S-methyl-5′-thioadenosine phosphorylase (MTAP), which had reduced expression in PV patients’ erythroblasts. Furthermore, MTAP knockdown in normal erythroblasts was shown to enhance their proliferative capacity. Together, this study identifies differentially expressed proteins in erythroblasts of healthy subjects and those of PV patients, indicating that an alteration of protein expression in erythroblasts may be crucial to the pathology of PV.
Collapse
Affiliation(s)
- Chartsiam Tipgomut
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Archrob Khuhapinant
- Division of Haematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | | | - Saiphon Poldee
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Kate J Heesom
- Proteomics Facility, University of Bristol, Bristol, BS8 1TD, UK
| | - Chanatip Metheetrairut
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Orapan Sripichai
- National Institute of Health, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, 11000, Thailand
| | - Chalermchai Mitrpant
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Jan Frayne
- School of Biochemistry, Faculty of Life Sciences, University of Bristol, Bristol, BS81TD, UK
| | - Kongtana Trakarnsanga
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
11
|
Abstract
Myeloproliferative neoplasms (MPNs) are clonal hematopoietic stem cell (HSC) disorders with overproduction of mature myeloid blood cells, including essential thrombocythemia (ET), polycythemia vera (PV), and primary myelofibrosis (PMF). In 2005, several groups identified a single gain-of-function point mutation JAK2V617F in the majority of MPN patients. The JAK2V617F mutation confers cytokine independent proliferation to hematopoietic progenitor cells by constitutively activating canonical and non-canonical downstream pathways. In this chapter, we focus on (1) the regulation of JAK2, (2) the molecular mechanisms used by JAK2V617F to induce MPNs, (3) the factors that are involved in the phenotypic diversity in MPNs, and (4) the effects of JAK2V617F on hematopoietic stem cells (HSCs). The discovery of the JAK2V617F mutation led to a comprehensive understanding of MPN; however, the question still remains about how one mutation can give rise to three distinct disease entities. Various mechanisms have been proposed, including JAK2V617F allele burden, differential STAT signaling, and host genetic modifiers. In vivo modeling of JAK2V617F has dramatically enhanced the understanding of the pathophysiology of the disease and provided the pre-clinical platform. Interestingly, most of these models do not show an increased hematopoietic stem cell self-renewal and function compared to wildtype controls, raising the question of whether JAK2V617F alone is sufficient to give a clonal advantage in MPN patients. In addition, the advent of modern sequencing technologies has led to a broader understanding of the mutational landscape and detailed JAK2V617F clonal architecture in MPN patients.
Collapse
|
12
|
Fahrmann JF, Irajizad E, Kobayashi M, Vykoukal J, Dennison JB, Murage E, Wu R, Long JP, Do KA, Celestino J, Lu KH, Lu Z, Bast RC, Hanash S. A MYC-Driven Plasma Polyamine Signature for Early Detection of Ovarian Cancer. Cancers (Basel) 2021; 13:913. [PMID: 33671595 PMCID: PMC7927060 DOI: 10.3390/cancers13040913] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/02/2021] [Accepted: 02/02/2021] [Indexed: 12/31/2022] Open
Abstract
MYC is an oncogenic driver in the pathogenesis of ovarian cancer. We previously demonstrated that MYC regulates polyamine metabolism in triple-negative breast cancer (TNBC) and that a plasma polyamine signature is associated with TNBC development and progression. We hypothesized that a similar plasma polyamine signature may associate with ovarian cancer (OvCa) development. Using mass spectrometry, four polyamines were quantified in plasma from 116 OvCa cases and 143 controls (71 healthy controls + 72 subjects with benign pelvic masses) (Test Set). Findings were validated in an independent plasma set from 61 early-stage OvCa cases and 71 healthy controls (Validation Set). Complementarity of polyamines with CA125 was also evaluated. Receiver operating characteristic area under the curve (AUC) of individual polyamines for distinguishing cases from healthy controls ranged from 0.74-0.88. A polyamine signature consisting of diacetylspermine + N-(3-acetamidopropyl)pyrrolidin-2-one in combination with CA125 developed in the Test Set yielded improvement in sensitivity at >99% specificity relative to CA125 alone (73.7% vs 62.2%; McNemar exact test 2-sided P: 0.019) in the validation set and captured 30.4% of cases that were missed with CA125 alone. Our findings reveal a MYC-driven plasma polyamine signature associated with OvCa that complemented CA125 in detecting early-stage ovarian cancer.
Collapse
Affiliation(s)
- Johannes F. Fahrmann
- Department of Clinical Cancer Prevention, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA; (J.F.F.); (M.K.); (J.V.); (J.B.D.); (E.M.); (R.W.)
| | - Ehsan Irajizad
- Department of Biostatistics, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA; (E.I.); (J.P.L.); (K.-A.D.)
| | - Makoto Kobayashi
- Department of Clinical Cancer Prevention, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA; (J.F.F.); (M.K.); (J.V.); (J.B.D.); (E.M.); (R.W.)
| | - Jody Vykoukal
- Department of Clinical Cancer Prevention, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA; (J.F.F.); (M.K.); (J.V.); (J.B.D.); (E.M.); (R.W.)
| | - Jennifer B. Dennison
- Department of Clinical Cancer Prevention, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA; (J.F.F.); (M.K.); (J.V.); (J.B.D.); (E.M.); (R.W.)
| | - Eunice Murage
- Department of Clinical Cancer Prevention, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA; (J.F.F.); (M.K.); (J.V.); (J.B.D.); (E.M.); (R.W.)
| | - Ranran Wu
- Department of Clinical Cancer Prevention, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA; (J.F.F.); (M.K.); (J.V.); (J.B.D.); (E.M.); (R.W.)
| | - James P. Long
- Department of Biostatistics, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA; (E.I.); (J.P.L.); (K.-A.D.)
| | - Kim-Anh Do
- Department of Biostatistics, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA; (E.I.); (J.P.L.); (K.-A.D.)
| | - Joseph Celestino
- Department of Gynecological Oncology and Reproductive Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA; (J.C.); (K.H.L.); (Z.L.)
| | - Karen H. Lu
- Department of Gynecological Oncology and Reproductive Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA; (J.C.); (K.H.L.); (Z.L.)
| | - Zhen Lu
- Department of Gynecological Oncology and Reproductive Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA; (J.C.); (K.H.L.); (Z.L.)
| | - Robert C. Bast
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA;
| | - Samir Hanash
- Department of Clinical Cancer Prevention, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA; (J.F.F.); (M.K.); (J.V.); (J.B.D.); (E.M.); (R.W.)
| |
Collapse
|
13
|
Fahrmann JF, Vykoukal J, Fleury A, Tripathi S, Dennison JB, Murage E, Wang P, Yu CY, Capello M, Creighton CJ, Do KA, Long JP, Irajizad E, Peterson C, Katayama H, Disis ML, Arun B, Hanash S. Association Between Plasma Diacetylspermine and Tumor Spermine Synthase With Outcome in Triple-Negative Breast Cancer. J Natl Cancer Inst 2021; 112:607-616. [PMID: 31503278 DOI: 10.1093/jnci/djz182] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 07/30/2019] [Accepted: 09/05/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND MYC is an oncogenic driver of development and progression in triple-negative breast cancer (TNBC). Ornithine decarboxylase, the rate-limiting enzyme in polyamine metabolism, is a transcriptional target of MYC. We therefore hypothesized that a plasma polyamine signature may be predictive of TNBC development and progression. METHODS Using liquid chromatography mass spectrometry, polyamine levels were determined in plasma samples from newly diagnosed patients with TNBC (n = 87) and cancer-free controls (n = 115). Findings were validated in plasma samples from an independent prospective cohort of 54 TNBC, 55 estrogen receptor negative (ER-) and progesterone receptor negative (PR-) and HER2 positive (HER2+), and 73 ER+ case patients, and 30 cancer-free control subjects. Gene expression data and clinical data for 921 and 2359 breast cancer tumors were obtained from The Cancer Genome Atlas repository and the Oncomine database, respectively. Relationships between plasma diacetylspermine (DAS) and tumor spermine synthase (SMS) mRNA expression with metastasis-free survival and overall survival were determined using Cox proportional hazard models; Fisher exact tests were used to assess risk of distant metastasis in relation to tumor SMS mRNA expression. RESULTS An increase in plasma DAS, a catabolic product of spermine mediated through SMS, was observed in the TNBC subtype of breast cancer. Plasma levels of DAS in TNBC associated with increased risk of metastasis (plasma DAS value ≥ 1.16, hazard ratio = 3.06, 95% confidence interval [CI] = 1.15 to 8.13, two-sided P = .03). SMS mRNA expression in TNBC tumor tissue was also found to be predictive of poor overall survival (top 25th percentile hazard ratio = 2.06, 95% CI = 1.04 to 4.08, one-sided P = .04) and increased risk of distant metastasis in TNBC (comparison of lowest SMS quartile [reference] to highest SMS quartile relative risk = 1.90, 95% CI = 0.97 to 4.06, one-sided Fisher exact test P=.03). CONCLUSIONS Metabolomic profiling identified plasma DAS as a predictive marker for TNBC progression and metastasis.
Collapse
Affiliation(s)
| | | | | | - Satyendra Tripathi
- Departments of Clinical Cancer Prevention.,Department of Biochemistry, AIIMS Nagpur, Nagpur, Maharashtra, India
| | | | | | - Peng Wang
- Departments of Clinical Cancer Prevention
| | | | | | - Chad J Creighton
- Bioinformatics and Computational Biology.,Department of Medicine and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston
| | | | | | | | | | | | - Mary L Disis
- University of Texas MD Anderson Cancer Center, Houston, TX; University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | | |
Collapse
|
14
|
Bhoopalan SV, Huang LJS, Weiss MJ. Erythropoietin regulation of red blood cell production: from bench to bedside and back. F1000Res 2020; 9:F1000 Faculty Rev-1153. [PMID: 32983414 PMCID: PMC7503180 DOI: 10.12688/f1000research.26648.1] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/04/2020] [Indexed: 12/18/2022] Open
Abstract
More than 50 years of efforts to identify the major cytokine responsible for red blood cell (RBC) production (erythropoiesis) led to the identification of erythropoietin (EPO) in 1977 and its receptor (EPOR) in 1989, followed by three decades of rich scientific discovery. We now know that an elaborate oxygen-sensing mechanism regulates the production of EPO, which in turn promotes the maturation and survival of erythroid progenitors. Engagement of the EPOR by EPO activates three interconnected signaling pathways that drive RBC production via diverse downstream effectors and simultaneously trigger negative feedback loops to suppress signaling activity. Together, the finely tuned mechanisms that drive endogenous EPO production and facilitate its downstream activities have evolved to maintain RBC levels in a narrow physiological range and to respond rapidly to erythropoietic stresses such as hypoxia or blood loss. Examination of these pathways has elucidated the genetics of numerous inherited and acquired disorders associated with deficient or excessive RBC production and generated valuable drugs to treat anemia, including recombinant human EPO and more recently the prolyl hydroxylase inhibitors, which act partly by stimulating endogenous EPO synthesis. Ongoing structure-function studies of the EPOR and its essential partner, tyrosine kinase JAK2, suggest that it may be possible to generate new "designer" drugs that control selected subsets of cytokine receptor activities for therapeutic manipulation of hematopoiesis and treatment of blood cancers.
Collapse
Affiliation(s)
- Senthil Velan Bhoopalan
- Department of Hematology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, MS #355, Memphis, TN, 38105, USA
| | - Lily Jun-shen Huang
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | - Mitchell J. Weiss
- Department of Hematology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, MS #355, Memphis, TN, 38105, USA
| |
Collapse
|
15
|
Hautin M, Mornet C, Chauveau A, Bernard DG, Corcos L, Lippert E. Splicing Anomalies in Myeloproliferative Neoplasms: Paving the Way for New Therapeutic Venues. Cancers (Basel) 2020; 12:E2216. [PMID: 32784800 PMCID: PMC7464941 DOI: 10.3390/cancers12082216] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/30/2020] [Accepted: 08/05/2020] [Indexed: 02/06/2023] Open
Abstract
Since the discovery of spliceosome mutations in myeloid malignancies, abnormal pre-mRNA splicing, which has been well studied in various cancers, has attracted novel interest in hematology. However, despite the common occurrence of spliceosome mutations in myelo-proliferative neoplasms (MPN), not much is known regarding the characterization and mechanisms of splicing anomalies in MPN. In this article, we review the current scientific literature regarding "splicing and myeloproliferative neoplasms". We first analyse the clinical series reporting spliceosome mutations in MPN and their clinical correlates. We then present the current knowledge about molecular mechanisms by which these mutations participate in the pathogenesis of MPN or other myeloid malignancies. Beside spliceosome mutations, splicing anomalies have been described in myeloproliferative neoplasms, as well as in acute myeloid leukemias, a dreadful complication of these chronic diseases. Based on splicing anomalies reported in chronic myelogenous leukemia as well as in acute leukemia, and the mechanisms presiding splicing deregulation, we propose that abnormal splicing plays a major role in the evolution of myeloproliferative neoplasms and may be the target of specific therapeutic strategies.
Collapse
Affiliation(s)
- Marie Hautin
- Inserm, Univ Brest, EFS, UMR 1078, GGB, F-29200 Brest, France; (M.H.); (A.C.); (D.G.B.); (L.C.)
| | - Clélia Mornet
- Laboratoire d’Hématologie, CHU de Brest, F-29200 Brest, France;
| | - Aurélie Chauveau
- Inserm, Univ Brest, EFS, UMR 1078, GGB, F-29200 Brest, France; (M.H.); (A.C.); (D.G.B.); (L.C.)
- Laboratoire d’Hématologie, CHU de Brest, F-29200 Brest, France;
| | - Delphine G. Bernard
- Inserm, Univ Brest, EFS, UMR 1078, GGB, F-29200 Brest, France; (M.H.); (A.C.); (D.G.B.); (L.C.)
| | - Laurent Corcos
- Inserm, Univ Brest, EFS, UMR 1078, GGB, F-29200 Brest, France; (M.H.); (A.C.); (D.G.B.); (L.C.)
| | - Eric Lippert
- Inserm, Univ Brest, EFS, UMR 1078, GGB, F-29200 Brest, France; (M.H.); (A.C.); (D.G.B.); (L.C.)
- Laboratoire d’Hématologie, CHU de Brest, F-29200 Brest, France;
| |
Collapse
|
16
|
Dong Y, Tu R, Liu H, Qing G. Regulation of cancer cell metabolism: oncogenic MYC in the driver's seat. Signal Transduct Target Ther 2020; 5:124. [PMID: 32651356 PMCID: PMC7351732 DOI: 10.1038/s41392-020-00235-2] [Citation(s) in RCA: 178] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/31/2022] Open
Abstract
Cancer cells must rewire cellular metabolism to satisfy the demands of unbridled growth and proliferation. As such, most human cancers differ from normal counterpart tissues by a plethora of energetic and metabolic reprogramming. Transcription factors of the MYC family are deregulated in up to 70% of all human cancers through a variety of mechanisms. Oncogenic levels of MYC regulates almost every aspect of cellular metabolism, a recently revisited hallmark of cancer development. Meanwhile, unrestrained growth in response to oncogenic MYC expression creates dependency on MYC-driven metabolic pathways, which in principle provides novel targets for development of effective cancer therapeutics. In the current review, we summarize the significant progress made toward understanding how MYC deregulation fuels metabolic rewiring in malignant transformation.
Collapse
Affiliation(s)
- Yang Dong
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Frontier Science Center for Immunology & Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
| | - Rongfu Tu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Frontier Science Center for Immunology & Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
| | - Hudan Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Frontier Science Center for Immunology & Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
| | - Guoliang Qing
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China. .,Frontier Science Center for Immunology & Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
17
|
Fan J, Feng Z, Chen N. Spermidine as a target for cancer therapy. Pharmacol Res 2020; 159:104943. [PMID: 32461185 DOI: 10.1016/j.phrs.2020.104943] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/16/2020] [Accepted: 05/19/2020] [Indexed: 12/13/2022]
Abstract
Spermidine, as a natural component from polyamine members, is originally isolated from semen and also existed in many natural plants, and can be responsible for cell growth and development in eukaryotes. The supplementation of spermidine can extend health and lifespan across species. Although the elevated levels of polyamines and the regulation of rate-limiting enzymes for polyamine metabolism have been identified as the biomarkers in many cancers, recent epidemiological data support that an increased uptake of spermidine as a caloric restriction mimic can reduce overall mortality associated with cancers. The possible mechanisms between spermidine and cancer development may be related to the precise regulation of polyamine metabolism, anti-cancer immunosurveillance, autophagy, and apoptosis. Increased intake of polyamine seems to suppress tumorigenesis, but appears to accelerate the growth of established tumors. Based on these observations and the absolute requirement for polyamines in tumor growth, spermidine could be a rational target for chemoprevention and clinical therapeutics of cancers.
Collapse
Affiliation(s)
- Jingjing Fan
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan 430079, China
| | - Ziyuan Feng
- Graduate School, Wuhan Sports University, Wuhan 430079, China
| | - Ning Chen
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan 430079, China.
| |
Collapse
|
18
|
Funakoshi-Tago M, Tsuruya R, Ueda F, Ishihara A, Kasahara T, Tamura H, Tago K. Tyrosine-phosphorylated SOCS3 negatively regulates cellular transformation mediated by the myeloproliferative neoplasm-associated JAK2 V617F mutant. Cytokine 2019; 123:154753. [PMID: 31255914 DOI: 10.1016/j.cyto.2019.154753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/31/2019] [Accepted: 06/07/2019] [Indexed: 01/30/2023]
Abstract
In the majority of myeloproliferative neoplasms (MPNs) patients, a point mutation, V617F has been found in Janus kinase 2 (JAK2) gene, and this JAK2 mutant provoked aberrant signaling pathway. In the current study, we found that suppressor of cytokine signaling proteins 3 (SOCS3) possessed the tumor suppressive activity against the JAK2 V617F mutant-provoked cellular transformation. The knockdown of SOCS3 increased the expression level of the JAK2 V617F mutant, which enhanced the activation of signaling mediators, including signal transducer and activator of transcription 3 and 5 (STAT3, STAT5) and extracellular signal-regulated kinase (ERK), and also increased of the proliferation rate and tumorigenesis activity of Ba/F3 cells expressing the JAK2 V617F mutant and erythropoietin receptor (EpoR). In contrast, the enforced expression of SOCS3 significantly inhibited the JAK2 V617F mutant-induced activation of downstream signaling molecules, cell proliferation, and tumorigenesis by down-regulating the expression level of the JAK2 V617F mutant. SOCS3 interacted with the JAK2V617F mutant through its SH2 domain and was phosphorylated at Tyr-204 and Tyr-221 in its SOCS box by the JAK2V617F mutant. SOCS3 mutants carrying a mutation in the SH2 domain (R71E) and a substitution at Tyr-221 (Y221F) failed to exert inhibitory effects on JAK2V617F mutant-induced cellular transformation and tumorigenesis. Collectively, these results imply that SOCS3 plays a negative role in the JAK2 V617F mutant-induced oncogenic signaling pathway through its SH2 domain and the phosphorylation of Tyr-221 in its SOCS box.
Collapse
Affiliation(s)
- Megumi Funakoshi-Tago
- Division of Hygienic Chemistry, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan.
| | - Rina Tsuruya
- Division of Hygienic Chemistry, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Fumihito Ueda
- Division of Hygienic Chemistry, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Aki Ishihara
- Division of Hygienic Chemistry, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Tadashi Kasahara
- International University of Health and Welfare, Graduate School, 1-3-3 Minamiaoyama, Minato-ku, Tokyo 107-0062, Japan
| | - Hiroomi Tamura
- Division of Hygienic Chemistry, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Kenji Tago
- Division of Structural Biochemistry, Department of Biochemistry, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi-ken 329-0498, Japan.
| |
Collapse
|
19
|
Abstract
Advances in our understanding of the metabolism and molecular functions of polyamines and their alterations in cancer have led to resurgence in the interest of targeting polyamine metabolism as an anticancer strategy. Increasing knowledge of the interplay between polyamine metabolism and other cancer-driving pathways, including the PTEN-PI3K-mTOR complex 1 (mTORC1), WNT signalling and RAS pathways, suggests potential combination therapies that will have considerable clinical promise. Additionally, an expanding number of promising clinical trials with agents targeting polyamines for both therapy and prevention are ongoing. New insights into molecular mechanisms linking dysregulated polyamine catabolism and carcinogenesis suggest additional strategies that can be used for cancer prevention in at-risk individuals. In addition, polyamine blocking therapy, a strategy that combines the inhibition of polyamine biosynthesis with the simultaneous blockade of polyamine transport, can be more effective than therapies based on polyamine depletion alone and may involve an antitumour immune response. These findings open up new avenues of research into exploiting aberrant polyamine metabolism for anticancer therapy.
Collapse
Affiliation(s)
- Robert A Casero
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.
| | - Tracy Murray Stewart
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Anthony E Pegg
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
20
|
Leifer BS, Doyle SK, Richters A, Evans HL, Koehler AN. An Array-Based Ligand Discovery Platform for Proteins With Short Half-Lives. Methods Enzymol 2018; 610:191-218. [PMID: 30390799 DOI: 10.1016/bs.mie.2018.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Many promising therapeutic protein targets were previously considered "undruggable" due to a deficit in structural information to guide drug design and/or a lack of an obvious binding pocket. Fortunately, array-based methods for evaluating protein binding against large chemical libraries, such as small-molecule microarray screening, have provided one of several emerging inroads to ligand discovery for these elusive targets. Despite the advance in the area of ligand discovery for poorly structured and intrinsically disordered proteins provided by array-based technologies involving cell lysates, the extension of this technology for screening proteins with short half-lives in physiologically relevant conformations has been technically challenging. In this chapter we present a protocol for leveraging in vitro translation strategies to enable array-based screening of short-lived proteins against large small-molecule libraries for ligand discovery.
Collapse
Affiliation(s)
- Becky S Leifer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States; Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States; The Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Shelby K Doyle
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States; Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States; The Broad Institute of MIT and Harvard, Cambridge, MA, United States; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - André Richters
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States; Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States; The Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Helen L Evans
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States; Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States; The Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Angela N Koehler
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States; Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States; The Broad Institute of MIT and Harvard, Cambridge, MA, United States; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States.
| |
Collapse
|
21
|
Pearson S, Williamson AJK, Blance R, Somervaille TCP, Taylor S, Azadbakht N, Whetton AD, Pierce A. Proteomic analysis of JAK2V617F-induced changes identifies potential new combinatorial therapeutic approaches. Leukemia 2017; 31:2717-2725. [PMID: 28533538 PMCID: PMC5729335 DOI: 10.1038/leu.2017.143] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/06/2017] [Accepted: 04/25/2017] [Indexed: 01/02/2023]
Abstract
In excess of 90% of patients with polycythaemia vera (PV) express a mutated form of Janus kinase 2 (JAK2), JAK2V617F. Such aberrant proteins offer great potential for the treatment of these diseases; however, inhibitors to JAK2 have had limited success in the clinic in terms of curing the disease. To understand the effects of this oncogene in haematopoietic cells with the aim of improving treatment strategies, we undertook a systematic evaluation of the effects of JAK2V617F expression using proteomics. The effects of JAK2V617F on over 5000 proteins and 2000 nuclear phosphopeptide sites were relatively quantified using either SILAC or eight-channel iTRAQ mass spectrometry. Pathway analysis of the proteins identified as changing indicated disruption to the p53 and MYC signalling pathways. These changes were confirmed using orthogonal approaches. The insight gained from this proteomic analysis led to the formation of hypothesis-driven analysis on inhibitor-mediated effects on primary cells from patients with a JAK2V617F mutation. Simultaneous inhibition of MYC and upregulation of p53 led to the preferential extinction of JAK2V617F-positive CD34+ cells, illustrating a potential therapeutic benefit from combined targeting of p53 and MYC.
Collapse
Affiliation(s)
- S Pearson
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, University of Manchester, Wolfson Molecular Imaging Centre, Manchester, UK
| | - A J K Williamson
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, University of Manchester, Wolfson Molecular Imaging Centre, Manchester, UK
| | - R Blance
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, University of Manchester, Wolfson Molecular Imaging Centre, Manchester, UK
| | - T C P Somervaille
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| | - S Taylor
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, University of Manchester, Wolfson Molecular Imaging Centre, Manchester, UK
| | - N Azadbakht
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, University of Manchester, Wolfson Molecular Imaging Centre, Manchester, UK
| | - A D Whetton
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, University of Manchester, Wolfson Molecular Imaging Centre, Manchester, UK
- Stoller Biomarker Discovery Centre, University of Manchester, Manchester, UK
| | - A Pierce
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, University of Manchester, Wolfson Molecular Imaging Centre, Manchester, UK
| |
Collapse
|
22
|
Dejure FR, Eilers M. MYC and tumor metabolism: chicken and egg. EMBO J 2017; 36:3409-3420. [PMID: 29127156 DOI: 10.15252/embj.201796438] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 09/16/2017] [Accepted: 09/28/2017] [Indexed: 12/17/2022] Open
Abstract
Transcription factors of the MYC family are deregulated in the majority of all human cancers. Oncogenic levels of MYC reprogram cellular metabolism, a hallmark of cancer development, to sustain the high rate of proliferation of cancer cells. Conversely, cells need to modulate MYC function according to the availability of nutrients, in order to avoid a metabolic collapse. Here, we review recent evidence that the multiple interactions of MYC with cell metabolism are mutual and review mechanisms that control MYC levels and function in response to metabolic stress situations. The main hypothesis we put forward is that regulation of MYC levels is an integral part of the adaptation of cells to nutrient deprivation. Since such mechanisms would be particularly relevant in tumor cells, we propose that-in contrast to growth factor-dependent controls-they are not disrupted during tumorigenesis and that maintaining flexibility of expression is integral to MYC's oncogenic function.
Collapse
Affiliation(s)
- Francesca R Dejure
- Theodor Boveri Institute and Comprehensive Cancer Center Mainfranken, Biocenter, University of Würzburg, Würzburg, Germany
| | - Martin Eilers
- Theodor Boveri Institute and Comprehensive Cancer Center Mainfranken, Biocenter, University of Würzburg, Würzburg, Germany
| |
Collapse
|
23
|
Funakoshi-Tago M, Moriwaki T, Ueda F, Tamura H, Kasahara T, Tago K. Phosphorylated CIS suppresses the Epo or JAK2 V617F mutant-triggered cell proliferation through binding to EpoR. Cell Signal 2017; 31:41-57. [DOI: 10.1016/j.cellsig.2016.12.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 11/21/2016] [Accepted: 12/23/2016] [Indexed: 12/01/2022]
|
24
|
Xu L, Long J, Wang P, Liu K, Mai L, Guo Y. Association between the ornithine decarboxylase G316A polymorphism and breast cancer survival. Oncol Lett 2015; 10:485-491. [PMID: 26171056 DOI: 10.3892/ol.2015.3201] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 02/19/2015] [Indexed: 11/05/2022] Open
Abstract
Ornithine decarboxylase (ODC) is a significant rate-limiting enzyme in polyamine synthesis, required for normal cell growth, and is highly expressed in various malignancies, including colorectal and breast cancer. In the present study, the associations between the ODC G316A single nucleotide polymorphism (SNP) and breast cancer-specific survival were investigated. In addition, the functional effects of this SNP were examined in the MCF-7 human breast cancer cell line. The present study recruited 300 stage I-III breast cancer cases, which were diagnosed at the Affiliated Cancer Hospital of Zhengzhou University (Zhengzhou, China) between 2002 and 2003, with follow-up visits conducted until May 2013. ODC G316A was genotyped (ODC GG vs. ODC AG/AA) in the 300 cases and the association of the genotypes with cancer-specific survival was analyzed. In the MCF-7 cell line, the ODC allele-specific binding of E-box transcription factors was determined using western blot and chromatin immunoprecipitation assays. Survival differences were observed between the two genotypes: Compared with the ODC GG genotype, patients with ODC GA/AA exhibited significantly higher survival rates (P<0.05). In cultured cells, the ODC SNP, which is flanked by two E-boxes, appeared to predict ODC promoter activity. Furthermore, the E-box activator c-MYC and repressor MAX interactor 1 were found to preferentially bind to ODC minor A-alleles compared with major G-alleles, in cultured MCF-7 cells. In conclusion, the results of the current study suggest that the regulation of ODC may affect survival in breast cancer patients and indicate a model in which the ODC SNP may be protective for breast adenoma recurrence and detrimental for survival following a diagnosis of breast cancer.
Collapse
Affiliation(s)
- Linping Xu
- Department of Scientific Research and Foreign Affairs, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Jianping Long
- Department of Breast Surgery, Maternity and Child-Care Hospital of Gansu Province, Lanzhou, Gansu 730050, P.R. China
| | - Peng Wang
- Henan Key Laboratory of Tumor Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Kangdong Liu
- Department of Scientific Research and Foreign Affairs, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Ling Mai
- Department of Scientific Research and Foreign Affairs, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Yongjun Guo
- Department of Scientific Research and Foreign Affairs, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| |
Collapse
|
25
|
Deficiency in WT1-targeting microRNA-125a leads to myeloid malignancies and urogenital abnormalities. Oncogene 2015; 35:1003-14. [PMID: 25961914 DOI: 10.1038/onc.2015.154] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 03/23/2015] [Accepted: 04/03/2015] [Indexed: 11/08/2022]
Abstract
The Wilms' tumor gene WT1 is overexpressed in leukemia and solid tumors and has an oncogenic role in leukemogenesis and tumorigenesis. However, precise regulatory mechanisms of WT1 overexpression remain undetermined. In the present study, microRNA-125a (miR-125a) was identified as a miRNA that suppressed WT1 expression via binding to the WT1-3'UTR. MiR-125a knockout mice overexpressed WT1, developed myeloproliferative disorder (MPD) characterized by expansion of myeloid cells in bone marrow (BM), spleen and peripheral blood, and displayed urogenital abnormalities. Silencing of WT1 expression in hematopoietic stem/progenitor cells of miR-125a knockout MPD mice by short-hairpin RNA inhibited myeloid colony formation in vitro. Furthermore, the incidence and severity of MPD were lower in miR-125a (-/-) mice than in miR-125a (+/-) mice, indicating the operation of compensatory mechanisms for the complete loss of miR-125a. To elucidate the compensatory mechanisms, miRNA array was performed. MiR-486 was occasionally induced in compete loss of miR-125a and inhibited WT1 expression instead of miR-125a, resulting in the cancellation of MPD occurrence. These results showed for the first time the post-transcriptional regulatory mechanisms of WT1 by both miR-125a and miR-486 and should contribute to the elucidation of mechanisms of normal hematopoiesis and kidney development.
Collapse
|
26
|
Kasahara T. [Study of cytokine signaling: the quest for immunomodulatory drugs interacting with cytokine production and activity]. YAKUGAKU ZASSHI 2015; 135:431-47. [PMID: 25759052 DOI: 10.1248/yakushi.14-00237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
I have been engaged in research and education in the fields of immunology and biochemistry at a medical college and college of pharmacy for 40 years. The original reasons why I began studying cytokines and some of the interests that have motivated me to continue working in the field of cytokine research are described: 1) the roles of cytokines in various immunological and inflammatory diseases (e.g., chemokines in bacterial infections and inflammatory diseases, particularly the role of interleukin-5 and eotaxins in eosinophilia); 2) the role of focal adhesion kinase in antiapoptosis and metastasis of melanoma; 3) recent findings on the role of JAK2/STAT pathways, particularly how JAK2V617F mutation induces dysregulated proliferation and tumorigenesis; and 4) the interactions of various chemical compounds and natural products in cytokine gene activation and signaling. Previous discoveries and published findings by my research group are described, along with comments and discussion pertaining to recent developments in the field.
Collapse
Affiliation(s)
- Tadashi Kasahara
- Graduate School, International University of Health and Welfare; 1-3-3 Minamiaoyama, Minato-ku, Tokyo 107-0062, Japan; Keio University Faculty of Pharmacy; 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan; Division of Inflammation Research, Jichi Medical University; 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| |
Collapse
|
27
|
Chen E, Mullally A. How does JAK2V617F contribute to the pathogenesis of myeloproliferative neoplasms? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2014; 2014:268-276. [PMID: 25696866 DOI: 10.1182/asheducation-2014.1.268] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
A decade on from the discovery of the JAK2V617F mutation in the majority of patients with myeloproliferative neoplasms (MPNs), JAK2V617F is now firmly installed in the hematology curriculum of medical students and the diagnostic-testing algorithm of clinicians. Furthermore, the oral JAK1/JAK2 inhibitor ruxolitinib, rationally designed to target activated JAK2 signaling in MPN, has been approved by the Food and Drug Administration (FDA) of the United States for the past 3 years for the treatment of intermediate- and advanced-phase myelofibrosis. Notwithstanding this, JAK2V617F continues to stimulate the MPN research community and novel insights into understanding the mechanisms by which JAK2V617F contributes to the pathogenesis of MPN are continually emerging. In this chapter, we focus on recent advances in 4 main areas: (1) the molecular processes coopted by JAK2V617F to induce MPN, (2) the role that JAK2V617F plays in phenotypic diversity in MPN, (3) the functional impact of JAK2V617F on hematopoietic stem cells, and (4) therapeutic strategies to target JAK2V617F. Although great strides have been made, significant deficits still exist in our understanding of the precise mechanisms by which JAK2V617F-mutant hematopoietic stem cells emerge and persist to engender clonal hematopoiesis in MPN and in developing strategies to preferentially target the JAK2V617F-mutant clone therapeutically. Critically, although myelofibrosis remains arguably the greatest clinical challenge in JAK2V617F-mediated MPN, the current understanding of myelofibrosis-specific disease biology remains quite rudimentary. Therefore, many important biological questions pertaining to JAK2V617F will continue to engage and challenge the MPN research community in the coming decade.
Collapse
Affiliation(s)
- Edwin Chen
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Ann Mullally
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
28
|
Effect of chemical modification on the ability of pyrrolidinium fullerene to induce apoptosis of cells transformed by JAK2 V617F mutant. Int Immunopharmacol 2014; 20:258-63. [PMID: 24631513 DOI: 10.1016/j.intimp.2014.02.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 02/26/2014] [Accepted: 02/26/2014] [Indexed: 11/22/2022]
Abstract
JAK2 V617F mutant, a gene responsible for human myeloproliferative neoplasms (MPNs), causes not only cellular transformation but also resistance to various anti-cancer drugs. We previously reported that pyrrolidinium fullerene markedly induced the apoptosis of JAK2 V617F mutant-induced transformed cells through the reduction of apoptosis signal-regulating kinase 1 (ASK1), following inhibition of the c-Jun N-terminal kinase (JNK) pathway. In the current study, we found that the replacement of the 2-hydrogen atom (H) or N-methyl group (CH3) by the butyl group (C4C9) caused the more than 3-fold potent cytotoxic effects on cells transformed by the JAK2 V617F mutant. Strikingly, these chemical modification of pyrrolidinium fullerene resulted in more marked reduction of ASK1 protein and a more potent inhibitory effect on the JNK signaling cascade. On the other hand, when modified with a longer alkyl group, the derivatives lacked their cytotoxicity. These observations clearly indicate that the modification of pyrrolidinium fullerene with a suitable length of alkyl group such as butyl group enhances its apoptotic effect through inhibition of the ASK1-MKK4/7-JNK pathway.
Collapse
|
29
|
Ueda F, Sumi K, Tago K, Kasahara T, Funakoshi-Tago M. Critical role of FANCC in JAK2 V617F mutant-induced resistance to DNA cross-linking drugs. Cell Signal 2013; 25:2115-24. [DOI: 10.1016/j.cellsig.2013.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 07/01/2013] [Indexed: 10/26/2022]
|