1
|
Lin CC, Hsu CL, Yao CY, Wang YH, Yuan CT, Kuo YY, Lee JY, Shih PT, Kao CJ, Chuang PH, Hsu YC, Hou HA, Chou WC, Tien HF. HOPX as a tumour-suppressive protein in T-cell acute lymphoblastic leukaemia. Br J Haematol 2025; 206:505-516. [PMID: 39737712 DOI: 10.1111/bjh.19965] [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: 09/02/2024] [Accepted: 12/16/2024] [Indexed: 01/01/2025]
Abstract
The homeodomain protein homeobox (HOPX), a multifaceted regulator of cellular functions and developmental processes, is predominantly expressed in stem cells across diverse tissues; it has also emerged as a tumour suppressor in various solid cancers. However, its role in haematological malignancies still remains undefined. This study aimed to elucidate its significance in T-cell acute lymphoblastic leukaemia (T-ALL). We firstly uncovered a novel link between reduced HOPX expression, its promoter hypermethylation and increased tumour burden in patients with T-ALL, suggesting its tumour-suppressive role. Next, we induced T-ALL by transducing intracellular NOTCH1 (ICN1) into mice with either conditional knock-in at the Rosa26 locus or knockout of Hopx. We found that T-ALL development was markedly accelerated and impeded in backgrounds with low and high Hopx expression respectively. Further analysis revealed Hopx's roles in modulating the Wnt-β-catenin pathway, a pivotal regulator of the downstream Myc signalling involved in T-ALL transformation and progression.
Collapse
Affiliation(s)
- Chien-Chin Lin
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chi-Yuan Yao
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Hung Wang
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chang-Tsu Yuan
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Pathology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Yuan-Yeh Kuo
- Tai-Cheng Stem Cell Therapy Center, National Taiwan University, Taipei, Taiwan
| | - Jhih-Yi Lee
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Pin-Tsen Shih
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chein-Jun Kao
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Po-Han Chuang
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yueh-Chwen Hsu
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsin-An Hou
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Chien Chou
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hwei-Fang Tien
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei, Taiwan
| |
Collapse
|
2
|
Zhang Z, Tang R, Zhu M, Zhu Z, Zhu J, Li H, Tong M, Li N, Huang J. Deciphering cell states and the cellular ecosystem to improve risk stratification in acute myeloid leukemia. Brief Bioinform 2024; 26:bbaf028. [PMID: 39865982 PMCID: PMC11770069 DOI: 10.1093/bib/bbaf028] [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: 10/26/2024] [Revised: 12/17/2024] [Accepted: 01/11/2025] [Indexed: 01/28/2025] Open
Abstract
Acute myeloid leukemia (AML) demonstrates significant cellular heterogeneity in both leukemic and immune cells, providing valuable insights into clinical outcomes. Here, we constructed an AML single-cell transcriptome atlas and proposed sciNMF workflow to systematically dissect underlying cellular heterogeneity. Notably, sciNMF identified 26 leukemic and immune cell states that linked to clinical variables, mutations, and prognosis. By examining the co-existence patterns among these cell states, we highlighted a unique AML cellular ecosystem (ACE) that signifies aberrant tumor milieu and poor survival, which is confirmed by public RNA-seq cohorts. We further developed the ACE signature (ACEsig), comprising 12 genes, which accurately predicts AML prognosis, and outperforms existing signatures. When applied to cytogenetically normal AML or intensively treated patients, the ACEsig continues to demonstrate strong performance. Our results demonstrate that large-scale systematic characterization of cellular heterogeneity has the potential to enhance our understanding of AML heterogeneity and contribute to more precise risk stratification strategy.
Collapse
Affiliation(s)
- Zheyang Zhang
- State Key Laboratory of Cellular Stress Biology, Xiang’an Hospital, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, No. 4221, Xiang’an South Road, Xiamen, Fujian 361102, China
- National Institute for Data Science in Health and Medicine, Xiamen University, No. 4221, Xiang’an South Road, Xiamen, Fujian 361102, China
| | - Ronghan Tang
- State Key Laboratory of Cellular Stress Biology, Xiang’an Hospital, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, No. 4221, Xiang’an South Road, Xiamen, Fujian 361102, China
| | - Ming Zhu
- State Key Laboratory of Cellular Stress Biology, Xiang’an Hospital, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, No. 4221, Xiang’an South Road, Xiamen, Fujian 361102, China
| | - Zhijuan Zhu
- Hematopoietic Stem Cell Transplantation Center, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Department of Hematology, Fujian Medical University Union Hospital, No. 29 Xinquan Street, Gulou District, Fuzhou 350001, China
| | - Jiali Zhu
- State Key Laboratory of Cellular Stress Biology, Xiang’an Hospital, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, No. 4221, Xiang’an South Road, Xiamen, Fujian 361102, China
| | - Hua Li
- Hematopoietic Stem Cell Transplantation Center, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Department of Hematology, Fujian Medical University Union Hospital, No. 29 Xinquan Street, Gulou District, Fuzhou 350001, China
- Department of Hematology and Rheumatology, The Second Affiliated Hospital of Xiamen Medical College, No. 566 Shengguang Road, Jimei District, Xiamen 361021, China
| | - Mengsha Tong
- State Key Laboratory of Cellular Stress Biology, Xiang’an Hospital, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, No. 4221, Xiang’an South Road, Xiamen, Fujian 361102, China
- National Institute for Data Science in Health and Medicine, Xiamen University, No. 4221, Xiang’an South Road, Xiamen, Fujian 361102, China
| | - Nainong Li
- Hematopoietic Stem Cell Transplantation Center, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Department of Hematology, Fujian Medical University Union Hospital, No. 29 Xinquan Street, Gulou District, Fuzhou 350001, China
- Translational Medicine Center on Hematology, Fujian Medical University, No. 29 Xinquan Street, Gulou District, Fuzhou 350001, China
| | - Jialiang Huang
- State Key Laboratory of Cellular Stress Biology, Xiang’an Hospital, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, No. 4221, Xiang’an South Road, Xiamen, Fujian 361102, China
- National Institute for Data Science in Health and Medicine, Xiamen University, No. 4221, Xiang’an South Road, Xiamen, Fujian 361102, China
| |
Collapse
|
3
|
Bury LAD, Fu S, Wynshaw-Boris A. Neuronal lineage tracing from progenitors in human cortical organoids reveals mechanisms of neuronal production, diversity, and disease. Cell Rep 2024; 43:114862. [PMID: 39395167 DOI: 10.1016/j.celrep.2024.114862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 08/14/2024] [Accepted: 09/25/2024] [Indexed: 10/14/2024] Open
Abstract
The contribution of progenitor subtypes to generating the billions of neurons produced during human cortical neurogenesis is not well understood. We developed the cortical organoid lineage-tracing (COR-LT) system for human cortical organoids. Differential fluorescent reporter activation in distinct progenitor cells leads to permanent reporter expression, enabling the progenitor cell lineage of neurons to be determined. Surprisingly, nearly all excitatory neurons produced in cortical organoids were generated indirectly from intermediate progenitor cells. Additionally, neurons of different progenitor lineages were transcriptionally distinct. Isogenic lines made from an autistic individual with and without a likely pathogenic CTNNB1 variant demonstrated that the variant substantially altered the proportion of neurons derived from specific progenitor cell lineages, as well as the lineage-specific transcriptional profiles of these neurons, suggesting a pathogenic mechanism for this mutation. These results suggest individual progenitor subtypes play roles in generating the diverse neurons of the human cerebral cortex.
Collapse
Affiliation(s)
- Luke A D Bury
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Shuai Fu
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Anthony Wynshaw-Boris
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
4
|
He F, Tu Y, Ni L. Research on the mechanism of HOPX-HDAC2 interaction inducing differentiation blockage in acute myeloid leukemia. Hematol Oncol 2024; 42:e3307. [PMID: 39243399 DOI: 10.1002/hon.3307] [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: 03/07/2024] [Revised: 06/20/2024] [Accepted: 08/06/2024] [Indexed: 09/09/2024]
Abstract
Homeodomain-only protein homeobox (HOPX) mainly exerts its transcriptional repression by physically sequestering the serum co-repressor and recruiting histone deacetylase (HDAC), possessing important potential as a prognostic gene in acute myeloid leukemia (AML). HDACs play crucial roles in cell growth, gene regulation, and metabolism, and they are also important factors in promoting AML progression. Therefore, this project attempts to investigate whether HOPX affects AML progression by interacting with HDAC2 protein. Bioinformatics analysis was employed to identify potential prognostic genes in AML. Flow cytometry and MTT assays were performed to analyze the cellular biological functions of the AML prognostic marker HOPX. The interaction network of HOPX was analyzed using the Search Tool for the Retrieval of Interacting Genes database, and the interaction between HOPX and HDAC2 was observed using endogenous and exogenous immunoprecipitation. HOPX is highly expressed in AML cells. Further research uncovered that low expression of HOPX can repress the proliferation activity, anti-apoptotic ability, and differentiation blockage of AML cells. Moreover, mechanistically, HOPX induced AML differentiation blockage and malignant progression through interaction with HDAC. HOPX can serve as a prognostic marker for AML and can interact with HDAC2 to induce AML differentiation blockage and malignant progression.
Collapse
Affiliation(s)
- Fang He
- Department of Hematology, Jinhua Municipal Central Hospital, Jinhua, China
| | - Yan Tu
- Department of Hematology, Jinhua Municipal Central Hospital, Jinhua, China
| | - Lihong Ni
- Department of Hematology, Jinhua Municipal Central Hospital, Jinhua, China
| |
Collapse
|
5
|
Lin C, Yao C, Wang Y, Hsu Y, Yuan C, Chen T, Hsu C, Lee S, Lee J, Shih P, Kao C, Chuang P, Kuo Y, Hou H, Chou W, Tien H. IDH2 mutation accelerates TPO-induced myelofibrosis with enhanced S100a8/a9 and NFκB signaling in vivo. EJHAEM 2024; 5:738-748. [PMID: 39157630 PMCID: PMC11327712 DOI: 10.1002/jha2.983] [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: 02/29/2024] [Revised: 06/23/2024] [Accepted: 07/08/2024] [Indexed: 08/20/2024]
Abstract
Introduction IDH2 mutation is an unfavorable prognostic factor in patients with primary myelofibrosis (PMF) but its effect on myelofibrosis (MF) remains largely unclear. Methods In this study, we aimed to elucidate the roles of IDH2 mutation in the development and progression of MF by transcriptomic and molecular techniques using the Idh2 R172K transgenic mice. Results We found that thrombopoietin (TPO)-overexpressed Idh2 R172K (Idh2 R172K + TPO) mice had accelerated progression to MF, compared with TPO-overexpressed Idh2-wild (WT + TPO) mice, showing activation of multiple inflammatory pathways, among which nuclear factor κB (NFκB) was the most significantly enhanced. Single-cell transcriptomes of the marrow cells in early MF showed that S100a8/a9 expression was mainly confined to neutrophil progenitors in the WT + TPO mice, but highly expressed in several types of myeloid precursor cells, including the megakaryocyte progenitors in the Idh2 R172K + TPO group. Furthermore, Idh2 R172K mice at age of 18 months had larger spleens, increased S100a8/a9-Tlr4 expression, and elevated serum S100a8/a9 levels compared with WT mice. PMF patients with IDH2 mutations had higher bone marrow plasma S100A8/A9 levels than those without IDH2 mutations. Conclusion Overall, our findings showed that IDH2 mutation induced proinflammatory effects, which further exacerbated MF, as evidenced by the increase in S100a8/a9 levels and NFκB hyperactivation in Idh2 R172K + TPO mice.
Collapse
Affiliation(s)
- Chien‐Chin Lin
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
- Division of HematologyDepartment of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
- Graduate Institute of Clinical Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
| | - Chi‐Yuan Yao
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
- Division of HematologyDepartment of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Yu‐Hung Wang
- Division of HematologyDepartment of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Yueh‐Chwen Hsu
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Chang‐Tsu Yuan
- Graduate Institute of Clinical Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Department of PathologyGraduate Institute of OncologyCollege of MedicineNational Taiwan UniversityTaipeiTaiwan
- Department of PathologyNational Taiwan University Cancer CenterTaipeiTaiwan
| | - Tsung‐Chih Chen
- Graduate Institute of Clinical Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Division of Hematology and Medical OncologyDepartment of Internal MedicineTaichung Veterans General HospitalTaipeiTaiwan
| | - Chia‐Lang Hsu
- Department of Medical ResearchNational Taiwan University Cancer CenterTaipeiTaiwan
| | - Sze‐Hwei Lee
- Division of Cellular TherapyDepartment of Integrated Diagnostics and TherapeuticsNational Taiwan University HospitalTaipeiTaiwan
| | - Jhih‐Yi Lee
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Pin‐Tsen Shih
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Chein‐Jun Kao
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Po‐Han Chuang
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Yuan‐Yeh Kuo
- Tai‐Cheng Stem Cell Therapy CenterNational Taiwan UniversityTaipeiTaiwan
| | - Hsin‐An Hou
- Division of HematologyDepartment of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Wen‐Chien Chou
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
- Division of HematologyDepartment of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Hwei‐Fang Tien
- Division of HematologyDepartment of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
- Division of Hematology and Medical OncologyDepartment of Internal MedicineFar Eastern Memorial HospitalNew TaipeiTaiwan
| |
Collapse
|
6
|
Lin L, Yu H, Xie X, Lei Q, Chen X, Su X, Wang X, Zhang S, Yang W. Leveraging FAM features to predict the prognosis of LGG patients and immunotherapy outcome. Am J Cancer Res 2024; 14:2731-2754. [PMID: 39005680 PMCID: PMC11236777 DOI: 10.62347/gigo3446] [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: 03/12/2024] [Accepted: 05/27/2024] [Indexed: 07/16/2024] Open
Abstract
Heterogeneity at biological and transcriptomic levels poses a challenge in defining and typing low-grade glioma (LGG), leading to a critical need for specific molecular signatures to enhance diagnosis, therapy, and prognostic evaluation of LGG. This study focused on fatty acid metabolism (FAM) related genes and prognostic features to investigate the mechanisms and treatment strategies for LGG cell metastasis and invasion. By screening 158 FAM-related genes and clustering 512 LGG samples into two subtypes (C1 and C2), differential gene expression analysis and functional enrichment were performed. The immune cell scores and prognosis were compared between the two subtypes, with C1 showing poorer outcomes and higher immune scores. A four-gene signature (PHEX, SHANK2, HOPX, and LGALS1) was identified and validated across different datasets, demonstrating a stable predictive effect. Cellular experiments confirmed the roles of LGALS1 and HOPX in promoting tumor cell proliferation, migration, and invasion, while SHANK2 exhibited a suppressive effect. This four-gene signature based on FAM-related genes offers valuable insights for understanding the pathogenesis and clinical management of LGG.
Collapse
Affiliation(s)
- Liangbin Lin
- Department of Neurosurgery and Urology, Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University Chengdu 610014, Sichuan, The People's Republic of China
- Obesity and Metabolism Medicine-Engineering Integration Laboratory, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University Chengdu 610031, The People's Republic of China
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University Chengdu 610031, The People's Republic of China
| | - Hui Yu
- Department of Neurosurgery and Urology, Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University Chengdu 610014, Sichuan, The People's Republic of China
| | - Xuelu Xie
- Department of Ophthalmology, West China School of Public Health and West China Forth Hospital, Sichuan University Chengdu 610041, Sichuan, The People's Republic of China
| | - Qingqiang Lei
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University Chongqing 400000, The People's Republic of China
| | - Xuerui Chen
- Department of Neurosurgery and Urology, Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University Chengdu 610014, Sichuan, The People's Republic of China
| | - Xu Su
- Department of Neurosurgery and Urology, Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University Chengdu 610014, Sichuan, The People's Republic of China
- College of Medicine, Southwest Jiaotong University Chengdu 610031, Sichuan, The People's Republic of China
| | - Xiuxuan Wang
- Department of Research and Development, Beijing DCTY Biotech Co., Ltd. Beijing 102200, The People's Republic of China
| | - Sunfu Zhang
- Department of Neurosurgery and Urology, Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University Chengdu 610014, Sichuan, The People's Republic of China
| | - Wenyong Yang
- Department of Neurosurgery and Urology, Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University Chengdu 610014, Sichuan, The People's Republic of China
| |
Collapse
|
7
|
Wang YH, Orgueira AM, Lin CC, Yao CY, Lo MY, Tsai CH, de la Fuente Burguera A, Hou HA, Chou WC, Tien HF. Stellae-123 gene expression signature improved risk stratification in taiwanese acute myeloid leukemia patients. Sci Rep 2024; 14:11064. [PMID: 38744924 PMCID: PMC11094146 DOI: 10.1038/s41598-024-61022-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 04/30/2024] [Indexed: 05/16/2024] Open
Abstract
The European Leukemia Net recommendations provide valuable guidance in treatment decisions of patients with acute myeloid leukemia (AML). However, the genetic complexity and heterogeneity of AML are not fully covered, notwithstanding that gene expression analysis is crucial in the risk stratification of AML. The Stellae-123 score, an AI-based model that captures gene expression patterns, has demonstrated robust survival predictions in AML patients across four western-population cohorts. This study aims to evaluate the applicability of Stellae-123 in a Taiwanese cohort. The Stellae-123 model was applied to 304 de novo AML patients diagnosed and treated at the National Taiwan University Hospital. We find that the pretrained (BeatAML-based) model achieved c-indexes of 0.631 and 0.632 for the prediction of overall survival (OS) and relapse-free survival (RFS), respectively. Model retraining within our cohort further improve the cross-validated c-indexes to 0.667 and 0.667 for OS and RFS prediction, respectively. Multivariable analysis identify both pretrained and retrained models as independent prognostic biomarkers. We further show that incorporating age, Stellae-123, and ELN classification remarkably improves risk stratification, revealing c-indices of 0.73 and 0.728 for OS and RFS, respectively. In summary, the Stellae-123 gene expression signature is a valuable prognostic tool for AML patients and model retraining can improve the accuracy and applicability of the model in different populations.
Collapse
Affiliation(s)
- Yu-Hung Wang
- Division of Hematology, National Taiwan University Hospital, Taipei, Taiwan
| | - Adrián Mosquera Orgueira
- Department of Hematology, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- Group of Computational Hematology and Genomics (GrHeCo-Xen), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Chien-Chin Lin
- Division of Hematology, National Taiwan University Hospital, Taipei, Taiwan.
- Department of Laboratory Medicine, National Taiwan University Hospital, No. 7, Chung-Shan S. Rd., Taipei City, 10002, Taiwan.
| | - Chi-Yuan Yao
- Division of Hematology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, No. 7, Chung-Shan S. Rd., Taipei City, 10002, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Min-Yen Lo
- Division of Hematology, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Cheng-Hong Tsai
- Division of Hematology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Medical Education and Research, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | | | - Hsin-An Hou
- Division of Hematology, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Chien Chou
- Division of Hematology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, No. 7, Chung-Shan S. Rd., Taipei City, 10002, Taiwan
| | - Hwei-Fang Tien
- Division of Hematology, National Taiwan University Hospital, Taipei, Taiwan.
- Department of Internal Medicine, Far-Eastern Memorial Hospital, No. 7, Chung-Shan S. Rd., Taipei City, 10002, Taiwan.
| |
Collapse
|
8
|
Mumme H, Thomas BE, Bhasin SS, Krishnan U, Dwivedi B, Perumalla P, Sarkar D, Ulukaya GB, Sabnis HS, Park SI, DeRyckere D, Raikar SS, Pauly M, Summers RJ, Castellino SM, Wechsler DS, Porter CC, Graham DK, Bhasin M. Single-cell analysis reveals altered tumor microenvironments of relapse- and remission-associated pediatric acute myeloid leukemia. Nat Commun 2023; 14:6209. [PMID: 37798266 PMCID: PMC10556066 DOI: 10.1038/s41467-023-41994-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/25/2023] [Indexed: 10/07/2023] Open
Abstract
Acute myeloid leukemia (AML) microenvironment exhibits cellular and molecular differences among various subtypes. Here, we utilize single-cell RNA sequencing (scRNA-seq) to analyze pediatric AML bone marrow (BM) samples from diagnosis (Dx), end of induction (EOI), and relapse timepoints. Analysis of Dx, EOI scRNA-seq, and TARGET AML RNA-seq datasets reveals an AML blasts-associated 7-gene signature (CLEC11A, PRAME, AZU1, NREP, ARMH1, C1QBP, TRH), which we validate on independent datasets. The analysis reveals distinct clusters of Dx relapse- and continuous complete remission (CCR)-associated AML-blasts with differential expression of genes associated with survival. At Dx, relapse-associated samples have more exhausted T cells while CCR-associated samples have more inflammatory M1 macrophages. Post-therapy EOI residual blasts overexpress fatty acid oxidation, tumor growth, and stemness genes. Also, a post-therapy T-cell cluster associated with relapse samples exhibits downregulation of MHC Class I and T-cell regulatory genes. Altogether, this study deeply characterizes pediatric AML relapse- and CCR-associated samples to provide insights into the BM microenvironment landscape.
Collapse
Affiliation(s)
- Hope Mumme
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
| | - Beena E Thomas
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Swati S Bhasin
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Upaasana Krishnan
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Bhakti Dwivedi
- Department of Biostatistics and Bioinformatics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Pruthvi Perumalla
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Debasree Sarkar
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Gulay B Ulukaya
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
| | - Himalee S Sabnis
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Sunita I Park
- Department of Pathology, Children's Healthcare of Atlanta, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Deborah DeRyckere
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Sunil S Raikar
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Melinda Pauly
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Ryan J Summers
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Sharon M Castellino
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Daniel S Wechsler
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Christopher C Porter
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Douglas K Graham
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Manoj Bhasin
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA.
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA.
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
9
|
Deng C, Zeng T, Zhu P, Zhao S, Huang Z, Huang W, Zhang W, Huang X, Fu L. A novel 5-gene prognostic signature to improve risk stratification of cytogenetically normal acute myeloid leukemia. J Cancer Res Clin Oncol 2023; 149:10015-10025. [PMID: 37258721 DOI: 10.1007/s00432-023-04884-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: 04/11/2023] [Accepted: 05/19/2023] [Indexed: 06/02/2023]
Abstract
PURPOSE Prognostic prediction is a challenging task in cytogenetically normal acute myeloid leukemia (CN-AML) patients. In this study, we aimed at developing a novel prognostic signature to predict and stratify the survival of CN-AML patients. METHODS Using a training dataset (GSE12417), 5-gene prognostic signature was established to predict survival of CN-AML patients. The prognostic performance of this prognostic signature was further validated in testing dataset (TCGA CN-AML cohort) and validation dataset (GSE6891 CN-AML cohort). RESULTS In training, testing and validation datasets, the increased 5-gene risk score was significantly related with inferior overall survival (OS) of patients, and the area under the receiver operating characteristic curve (AUC) demonstrated that our prognostic signature had overall prediction accuracy. The excellent prognostic value of the 5-gene prognostic signature was also supported by the comparison with three previously proposed prognostic models. For the intermediate-risk CN-AML patients and the CN-AML patients with FLT3 or NPM1 mutation, our model could also well dichotomize them into two subgroups with distinct prognosis. Multivariate analysis demonstrated that 5-gene risk score was the only independent risk factor in TCGA CN-AML cohort. Nomogram including the 5-gene risk score performed well in predicting 1-year, 2-year and 3-year OS. CONCLUSION In summary, our novel 5-gene prognostic signature facilitated the improvement in risk stratification of CN-AML patients.
Collapse
Affiliation(s)
- Cong Deng
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
- Department of Clinical Laboratory, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
- Central Laboratory, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Tiansheng Zeng
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
- Central Laboratory, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Pei Zhu
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
- Central Laboratory, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Sijie Zhao
- The Second Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Zeyong Huang
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
- Central Laboratory, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Wenhui Huang
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
- Central Laboratory, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Wenjuan Zhang
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
- Central Laboratory, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Xiaojuan Huang
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
- Central Laboratory, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Lin Fu
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.
- Central Laboratory, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.
| |
Collapse
|
10
|
Wang L, Nie R, Zhang J, Cai J. scCapsNet-mask: an updated version of scCapsNet with extended applicability in functional analysis related to scRNA-seq data. BMC Bioinformatics 2022; 23:539. [PMID: 36510124 PMCID: PMC9743530 DOI: 10.1186/s12859-022-05098-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 12/03/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND With the rapid accumulation of scRNA-seq data, more and more automatic cell type identification methods have been developed, especially those based on deep learning. Although these methods have reached relatively high prediction accuracy, many issues still exist. One is the interpretability. The second is how to deal with the non-standard test samples that are not encountered in the training process. RESULTS Here we introduce scCapsNet-mask, an updated version of scCapsNet. The scCapsNet-mask provides a reasonable solution to the issues of interpretability and non-standard test samples. Firstly, the scCapsNet-mask utilizes a mask to ease the task of model interpretation in the original scCapsNet. The results show that scCapsNet-mask could constrain the coupling coefficients, and make a one-to-one correspondence between the primary capsules and type capsules. Secondly, the scCapsNet-mask can process non-standard samples more reasonably. In one example, the scCapsNet-mask was trained on the committed cells, and then tested on less differentiated cells as the non-standard samples. It could not only estimate the lineage bias of less differentiated cells, but also distinguish the development stages more accurately than traditional machine learning models. Therefore, the pseudo-temporal order of cells for each lineage could be established. Following these pseudo-temporal order, lineage specific genes exhibit a gradual increase expression pattern and stem cell associated genes exhibit a gradual decrease expression pattern. In another example, the scCapsNet-mask was trained on scRNA-seq data, and then used to assign cell type in spatial transcriptomics that may contain non-standard sample of doublets. The results show that the scCapsNet-mask not only restored the spatial map but also identified several non-standard samples of doublet. CONCLUSIONS The scCapsNet-mask offers a suitable solution to the challenge of interpretability and non-standard test samples. By adding a mask, it has the advantages of automatic processing and easy interpretation compared with the original scCapsNet. In addition, the scCapsNet-mask could more accurately reflect the composition of non-standard test samples than traditional machine learning methods. Therefore, it can extend its applicability in functional analysis, such as fate bias prediction in less differentiated cells and cell type assignment in spatial transcriptomics.
Collapse
Affiliation(s)
- Lifei Wang
- grid.413073.20000 0004 1758 9341Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China ,grid.464209.d0000 0004 0644 6935China National Center for Bioinformation, Beijing, 100101 China ,grid.9227.e0000000119573309Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Rui Nie
- grid.464209.d0000 0004 0644 6935China National Center for Bioinformation, Beijing, 100101 China ,grid.9227.e0000000119573309Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Jiang Zhang
- grid.20513.350000 0004 1789 9964School of Systems Science, Beijing Normal University, Beijing, 100875 China
| | - Jun Cai
- grid.464209.d0000 0004 0644 6935China National Center for Bioinformation, Beijing, 100101 China ,grid.9227.e0000000119573309Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| |
Collapse
|
11
|
Yao CY, Lin CC, Wang YH, Hsu CL, Kao CJ, Hou HA, Chou WC, Tien HF. The clinical and biological characterization of acute myeloid leukemia patients with S100A4 overexpression. J Formos Med Assoc 2022:S0929-6646(22)00422-3. [DOI: 10.1016/j.jfma.2022.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 10/31/2022] [Accepted: 11/07/2022] [Indexed: 11/24/2022] Open
|
12
|
Chanoch-Myers R, Wider A, Suva ML, Tirosh I. Elucidating the diversity of malignant mesenchymal states in glioblastoma by integrative analysis. Genome Med 2022; 14:106. [PMID: 36123598 PMCID: PMC9484143 DOI: 10.1186/s13073-022-01109-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 08/31/2022] [Indexed: 12/15/2022] Open
Abstract
Background Multiple glioblastoma studies have described a mesenchymal (MES) state, with each study defining the MES program by distinct sets of genes and highlighting distinct functional associations, including both immune activation and suppression. These variable descriptions complicate our understanding of the MES state and its implications. Here, we hypothesize that there is a range of glioma MES states, possibly reflecting distinct prior states in which a MES program can be induced, and/or distinct mechanisms that induce the MES states in those cells. Methods We integrated multiple published single-cell and bulk RNA sequencing datasets and MES signatures to define a core MES program that recurs across studies, as well as multiple function-specific MES signatures that vary across MES cells. We then examined the co-occurrence of these signatures and their associations with genetic and microenvironmental features. Results Based on co-occurrence of MES signatures, we found three main variants of MES states: hypoxia-related (MES-Hyp), astrocyte-related (MES-Ast), and an intermediate state. Notably, the MES states are differentially associated with genetic and microenvironmental features. MES-Hyp is preferentially associated with NF1 deletion, overall macrophage abundance, a high macrophage/microglia ratio, and M2-related macrophages, consistent with previous studies that associated MES with immune suppression. In contrast, MES-Ast is associated with T cell abundance and cytotoxicity, consistent with immune activation through expression of MHC-I/II. Conclusions Diverse MES states occur in glioblastoma. These states share a subset of core genes but differ primarily in their association with hypoxia vs. astrocytic expression programs, and with immune suppression vs. activation, respectively. Supplementary Information The online version contains supplementary material available at 10.1186/s13073-022-01109-8.
Collapse
Affiliation(s)
- Rony Chanoch-Myers
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Adi Wider
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Mario L Suva
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Itay Tirosh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
13
|
Lin SC, Yao CY, Hsu CA, Lin CT, Calkins MJ, Kuo YY, Tang JL, Tien HF, Wu SJ. Functional association of NR4A3 downregulation with impaired differentiation in myeloid leukemogenesis. Ann Hematol 2022; 101:2209-2218. [PMID: 36040481 PMCID: PMC9463347 DOI: 10.1007/s00277-022-04961-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 08/17/2022] [Indexed: 11/29/2022]
Abstract
The coincident downregulation of NR4A1 and NR4A3 has been implicated in myeloid leukemogenesis, but it remains unknown how these two genes function in myeloid cells and how their combined downregulation promotes myeloid leukemogenesis. Since NR4A1 abrogation is thought to confer a survival and proliferation advantage to myeloid cells, we hypothesized that downregulation of NR4A3 may have a complementary effect on myeloid cell differentiation. First, we tested the association between differentiation status of leukemic cells and NR4A3 expression using two large clinical datasets from patients with different acute myeloid leukemia (AML) subtypes. The analysis revealed a close association between differentiation status and different subtypes of AML Then, we probed the effects of differentiation-inducing treatments on NR4A3 expression and NR4A3 knockdown on cell differentiation using two myeloid leukemia cell lines. Differentiation-inducing treatments caused upregulation of NR4A3, while NR4A3 knockdown prevented differentiation in both cell lines. The cell culture findings were validated using samples from chronic myeloid leukemia (CML) patients at chronic, accelerated and blastic phases, and in acute promyelocytic leukemia (APL) patients before and after all trans-retinoic acid (ATRA)-based differentiation therapy. Progressive NR4A3 downregulation was coincident with impairments in differentiation in patients during progression to blastic phase of CML, and NR4A3 expression was increased in APL patients treated with ATRA-based differentiating therapy. Together, our findings demonstrate a tight association between impaired differentiation status and NR4A3 downregulation in myeloid leukemias, providing a plausible mechanistic explanation of how myeloid leukemogenesis might occur upon concurrent downregulation of NR4A1 and NR4A3.
Collapse
Affiliation(s)
- Shih-Chiang Lin
- Department of Internal Medicine, Far-Eastern Memorial Hospital, New Taipei City, Taiwan.,General Education Center, Lunghwa University of Science and Technology, Taoyuan City, Taiwan
| | - Chi-Yuan Yao
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei City, Taiwan.,Department of Internal Medicine, Zhongzheng Dist, National Taiwan University Hospital, No.7, Chung Shan S. Rd, Taipei City, 100225, Taiwan
| | - Cheng-An Hsu
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei City, Taiwan
| | - Chien-Ting Lin
- Department of Internal Medicine, Zhongzheng Dist, National Taiwan University Hospital, No.7, Chung Shan S. Rd, Taipei City, 100225, Taiwan.,Tai-Cheng Cell Therapy Center, National Taiwan University Cancer Center, Taipei City, Taiwan.,Pell Bio-Med Technology CO., LTD., Taipei City, Taiwan.,Department of Hematological Oncology, National Taiwan University Cancer Center, Taipei City, Taiwan
| | - Marcus J Calkins
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei City, Taiwan
| | - Yuan-Yeh Kuo
- Tai-Cheng Cell Therapy Center, National Taiwan University Cancer Center, Taipei City, Taiwan
| | - Jih-Luh Tang
- Department of Internal Medicine, Zhongzheng Dist, National Taiwan University Hospital, No.7, Chung Shan S. Rd, Taipei City, 100225, Taiwan.,Tai-Cheng Cell Therapy Center, National Taiwan University Cancer Center, Taipei City, Taiwan.,Department of Hematological Oncology, National Taiwan University Cancer Center, Taipei City, Taiwan
| | - Hwei-Fang Tien
- Department of Internal Medicine, Zhongzheng Dist, National Taiwan University Hospital, No.7, Chung Shan S. Rd, Taipei City, 100225, Taiwan
| | - Shang-Ju Wu
- Department of Internal Medicine, Zhongzheng Dist, National Taiwan University Hospital, No.7, Chung Shan S. Rd, Taipei City, 100225, Taiwan. .,Department of Hematological Oncology, National Taiwan University Cancer Center, Taipei City, Taiwan.
| |
Collapse
|
14
|
Bourque J, Kousnetsov R, Hawiger D. Roles of Hopx in the differentiation and functions of immune cells. Eur J Cell Biol 2022; 101:151242. [DOI: 10.1016/j.ejcb.2022.151242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/20/2022] [Accepted: 05/20/2022] [Indexed: 11/03/2022] Open
|
15
|
HOPX: A Unique Homeodomain Protein in Development and Tumor Suppression. Cancers (Basel) 2022; 14:cancers14112764. [PMID: 35681746 PMCID: PMC9179269 DOI: 10.3390/cancers14112764] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 12/05/2022] Open
Abstract
Simple Summary Homeobox (HOX) genes encode homeodomain proteins that regulate a wide range of molecular pathways. The homeodomain is highly conserved and binds to DNA. One exception is homeodomain-only protein (HOPX) that lacks DNA-binding capacity. HOPX plays a crucial role in development and its functional impairment is associated with a variety of diseases, including cancer. Loss of HOPX function occurs in a wide range of cancer types, where it functions as a tumor suppressor gene. Understanding the molecular mechanisms by which HOPX regulates carcinogenesis will likely lead to the development of new therapeutic approaches. Abstract Homeobox genes are master regulators of morphogenesis and differentiation by acting at the top of genetic hierarchies and their deregulation is associated with a variety of human diseases. They usually contain a highly conserved sequence that codes for the homeodomain of the protein, a specialized motif with three α helices and an N-terminal arm that aids in DNA binding. However, one homeodomain protein, HOPX, is unique among its family members in that it lacks the capacity to bind DNA and instead functions by interacting with transcriptional regulators. HOPX plays crucial roles in organogenesis and is expressed in both embryonic and adult stem cells. Loss of HOPX expression is common in cancer, where it functions primarily as a tumor suppressor gene. In this review, we describe the function of HOPX in development and discuss its role in carcinogenesis.
Collapse
|
16
|
Bourque J, Opejin A, Surnov A, Iberg CA, Gross C, Jain R, Epstein JA, Hawiger D. Landscape of Hopx expression in cells of the immune system. Heliyon 2021; 7:e08311. [PMID: 34805566 PMCID: PMC8590040 DOI: 10.1016/j.heliyon.2021.e08311] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/30/2021] [Accepted: 10/29/2021] [Indexed: 11/29/2022] Open
Abstract
Homeodomain only protein (Hopx) is a regulator of cell differentiation and function, and it has also emerged as a crucial marker of specific developmental and differentiation potentials. Hopx expression and functions have been identified in some stem cells, tumors, and in certain immune cells. However, expression of Hopx in immune cells remains insufficiently characterized. Here we report a comprehensive pattern of Hopx expression in multiple types of immune cells under steady state conditions. By utilizing single-cell RNA sequencing (scRNA-seq) and flow cytometric analysis, we characterize a constitutive expression of Hopx in specific subsets of CD4+ and CD8+ T cells and B cells, as well as natural killer (NK), NKT, and myeloid cells. In contrast, Hopx expression is not present in conventional dendritic cells and eosinophils. The utility of identifying expression of Hopx in immune cells may prove vital in delineating specific roles of Hopx under multiple immune conditions.
Collapse
Affiliation(s)
- Jessica Bourque
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63118, USA
| | - Adeleye Opejin
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63118, USA
| | - Alexey Surnov
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63118, USA
| | - Courtney A Iberg
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63118, USA
| | - Cindy Gross
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63118, USA
| | - Rajan Jain
- Department of Medicine, Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jonathan A Epstein
- Department of Medicine, Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Daniel Hawiger
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63118, USA
| |
Collapse
|
17
|
Wang YH, Lin CC, Yao CY, Hsu CL, Tsai CH, Hou HA, Chou WC, Tien HF. Immune signatures of bone marrow cells can independently predict prognosis in patients with myelodysplastic syndrome. Br J Haematol 2021; 196:156-168. [PMID: 34536013 DOI: 10.1111/bjh.17837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/24/2021] [Accepted: 09/02/2021] [Indexed: 01/17/2023]
Abstract
Increasing evidence supports the role of the immune microenvironment and associated signalling in the pathogenesis of myelodysplastic syndromes (MDS). Nevertheless, the clinical relevancy of immune signals in patients with MDS remains elusive. To address this, we used single-sample gene-set enrichment analysis to score immune signatures of bone marrow (BM) samples from 176 patients with primary MDS. Enhanced signatures of 'immature dendritic cells' and 'natural killer cells with cluster of differentiation (CD)56bright' were correlated with better overall survival (OS), whilst higher 'CD103+ signature' was associated with reduced survival. An MDS-Immune-Risk (MIR) scoring system was constructed based on the weighted sums derived from Cox regression analysis. High MIR scores were correlated with higher revised International Prognostic Scoring System (IPSS-R) scores and mutations in ASXL transcriptional regulator 1 (ASXL1), Runt-related transcription factor 1 (RUNX1), and tumour protein p53 (TP53). High-score patients had significantly inferior leukaemia-free survival (LFS) and OS than low-score patients. The prognostic significance of MIR scores for survival remained valid across IPSS-R subgroups and was validated in two independent cohorts. Multivariable analysis revealed that a higher MIR score was an independent adverse risk factor for LFS and OS. We further proposed a model with the combination of MIR score and gene mutations to be complementary to IPSS-R for the prognostication of LFS and OS of patients with MDS.
Collapse
Affiliation(s)
- Yu-Hung Wang
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Division of Haematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chien-Chin Lin
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Division of Haematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chi-Yuan Yao
- Division of Haematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Cheng-Hong Tsai
- Division of Haematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsin-An Hou
- Division of Haematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Chien Chou
- Division of Haematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hwei-Fang Tien
- Division of Haematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
18
|
Zhao H, Zhang X, Shi Z, Guo B, Zhang W, He K, Hu X, Shi S. Identification of a Prognostic Signature Model with Tumor Microenvironment for predicting Disease-free Survival after Radical Prostatectomy. J Cancer 2021; 12:2371-2384. [PMID: 33758613 PMCID: PMC7974886 DOI: 10.7150/jca.51173] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 01/18/2021] [Indexed: 12/24/2022] Open
Abstract
Background: The tumor microenvironment (TME) and immune checkpoint inhibitors have been shown to promote active immune responses through different mechanisms. We attempted to identify the important prognostic genes and prognostic characteristics related to TME in prostate cancer (PCa). Methods: The gene transcriptome profiles and clinical information of PCa patients were obtained from The Cancer Genome Atlas (TCGA) database, and the immune and stromal scores were calculated by the ESTIMATE algorithm. We evaluated the prognostic value of the risk score (RS) model based on univariate Cox analysis and least absolute shrinkage and selection operation (LASSO) Cox regression analysis and established a nomogram to predict disease-free survival (DFS) in PCa patients. The GSE70768 dataset was utilized for external validation. Twenty-two subsets of tumor-infiltrating immune cells were analyzed using the CIBERSORT algorithm. Results: In this study, the patients with higher immune/stromal scores were associated with a worse DFS, higher Gleason score, and higher pathological T stage. Based on the immune and stromal scores, 515 differentially expressed genes (DEGs) were identified. The univariate Cox and LASSO Cox regression models were employed to select 18 DEGs from 515 DEGs and construct an RS model. The DFS of the high-RS group was significantly lower than that of the low-RS group (P<0.001). The AUCs for the 1-year, 3-year and 5-year DFS rates in the RS model were 0.890, 0.877 and 0.841, respectively. A nomogram of DFS was established based on the RS and Gleason score, and the AUCs for the 1-year, 3-year and 5-year DFS rates in the nomogram were 0.907, 0.893, and 0.872, respectively. These results were further validated in the GSE70768 dataset. In addition, the proportion of Tregs was determined to be higher in high-RS patients (P<0.05), and the expression levels of five immune checkpoints (CTLA-4, PD-1, LAG-3, TIM-3 and TIGIT) were observed to be higher in high-RS patients (P<0.05). Conclusions: Our study established and validated an 18-gene prognostic signature model associated with TME, which might serve as a prognosis stratification tool to predict DFS in PCa patients after radical prostatectomy.
Collapse
Affiliation(s)
- Hao Zhao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Xuening Zhang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Zhan Shi
- Department of Medicine, Zhengzhou First People's Hospital, Zhengzhou 450004, China
| | - Bingxin Guo
- Department of Urology, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou 450002, China
| | - Wenli Zhang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Kun He
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Xueqi Hu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Songhe Shi
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
19
|
Abstract
In this issue of JEM, Louka et al. (https://doi.org/10.1084/jem.20180853) report that leukemia stem cells lie within the phenotypic hematopoietic stem cell and progenitor cell compartments in juvenile myelomonocytic leukemia (JMML). Furthermore, they identify several candidate biomarker/therapeutic targets, such as CD96 and SLC2A1, that are of translational significance in JMML.
Collapse
Affiliation(s)
- Sriram Sundaravel
- Albert Einstein College of Medicine—Montefiore Medical Center, Bronx, NY
| | - Ulrich Steidl
- Albert Einstein College of Medicine—Montefiore Medical Center, Bronx, NY
| |
Collapse
|
20
|
The role of HOPX in normal tissues and tumor progression. Biosci Rep 2020; 40:221873. [PMID: 31934721 PMCID: PMC6997107 DOI: 10.1042/bsr20191953] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 12/03/2019] [Accepted: 12/23/2019] [Indexed: 02/06/2023] Open
Abstract
The homeodomain-only protein homeobox (HOPX) as the smallest homeodomain protein, lacks certain conserved residues required for DNA binding. Through our literature search, we reviewed the current understandings of HOPX in normal tissues and tumor progression. HOPX was initially identified as a critical transcription factor in various normal tissues, which interacted with serum response factor (SRF) or other substance to regulate normal physiological function. However, HOPX is at a low expression or methylation level in tumors. These data indicated that HOPX may play a very important role in regulating differentiation phenotype and tumor suppressive function. We predicted the prognosis of HOPX in tumors from TCGA database and discussed the downstream genes of HOPX. To understand how HOPX is involved in the mechanisms between physical and pathological conditions could lead to novel therapeutic strategies for treatment.
Collapse
|
21
|
Hung SY, Lin CC, Hsu CL, Yao CY, Wang YH, Tsai CH, Hou HA, Chou WC, Tien HF. The expression levels of long non-coding RNA KIAA0125 are associated with distinct clinical and biological features in myelodysplastic syndromes. Br J Haematol 2020; 192:589-598. [PMID: 33249572 DOI: 10.1111/bjh.17231] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/20/2020] [Indexed: 12/16/2022]
Abstract
Long non-coding RNAs (lncRNAs) have important functions in cancer biology. Among them, lncRNA KIAA0125 is one of the genes proposed to play a critical role in leukaemia stem cell (LSC). In this study, we aimed to investigate the clinical relevance of the expression levels of lncRNA KIAA0125 in myelodysplastic syndromes (MDS), a disease with highly heterogeneous clinical and biological features. Using RNA arrays, we measured the expression of KIAA0125 in 176 primary MDS patients. We found that higher KIAA0125 expression was associated with higher risk MDS, based on the revised International Prognostic Scoring System (IPSS-R), mutations in ASXL1 and NRAS, and predicted poorer overall survival (OS) and leukaemia-free survival (LFS). Multivariate analysis revealed that higher KIAA0125 expression was an independent, unfavourable prognostic factor for OS and LFS, irrespective of IPSS-R and mutation status. Further global gene expression profile analysis suggested a close association of higher KIAA0125 expressions with LSC signatures. The expression of KIAA0125 may be a potential biomarker to guide the treatment choice in MDS patients, especially those with lower risk subtypes, in whom palliative treatment is usually used.
Collapse
Affiliation(s)
- Sheng-Yu Hung
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Chien-Chin Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei, Taiwan.,Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Chi-Yuan Yao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei, Taiwan.,Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Hung Wang
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Cheng-Hong Tsai
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsin-An Hou
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Chien Chou
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hwei-Fang Tien
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
22
|
Distinct clinical and biological characteristics of acute myeloid leukemia with higher expression of long noncoding RNA KIAA0125. Ann Hematol 2020; 100:487-498. [PMID: 33225420 PMCID: PMC7817567 DOI: 10.1007/s00277-020-04358-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022]
Abstract
Expression of long non-coding RNA KIAA0125 has been incorporated in various gene expression signatures for prognostic prediction in acute myeloid leukemia (AML) patients, yet its functions and clinical significance remain unclear. This study aimed to investigate the clinical and biological characteristics of AML bearing different levels of KIAA0125. We profiled KIAA0125 expression levels in bone marrow cells from 347 de novo AML patients and found higher KIAA0125 expression was closely associated with RUNX1 mutation, but inversely correlated with t(8;21) and t(15;17) karyotypes. Among the 227 patients who received standard chemotherapy, those with higher KIAA0125 expression had a lower complete remission rate, shorter overall survival (OS) and disease-free survival (DFS) than those with lower expression. The prognostic significance was validated in both TCGA and GSE12417 cohorts. Subgroup analyses showed that higher KIAA0125 expression also predicted shorter DFS and OS in patients with normal karyotype or non-M3 AML. In multivariable analysis, higher KIAA0125 expression remained an adverse risk factor independent of age, WBC counts, karyotypes, and mutation patterns. Bioinformatics analyses revealed that higher KIAA0125 expression was associated with hematopoietic and leukemic stem cell signatures and ATP-binding cassette transporters, two predisposing factors for chemoresistance.
Collapse
|
23
|
HSPG2 overexpression independently predicts poor survival in patients with acute myeloid leukemia. Cell Death Dis 2020; 11:492. [PMID: 32606327 PMCID: PMC7327006 DOI: 10.1038/s41419-020-2694-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 06/13/2020] [Accepted: 06/16/2020] [Indexed: 12/31/2022]
Abstract
Heparan sulfate proteoglycan 2 (HSPG2), also known as perlecan, is a large multi-domain extracellular matrix proteoglycan, which contributes to the invasion, metastasis and angiogenesis of solid tumor. However, very little is known about the effect of HSPG2 on acute myeloid leukemia (AML). This study aims to investigate the prognostic value of the HSPG2 gene in terms of overall survival and leukemia-free survival in patients with AML. Bone marrow mononuclear cells (BMMCs) from 4 AML patients and 3 healthy controls were processed for RNA-Sequencing (RNA-seq). The mRNA expression level of HSPG2 in BMMCs and CD34+ hematopoietic stem/progenitor cells (HSPC) obtained from enrolled participants and human leukemic cell lines was detected by RT-qPCR. Then the correlations between the expression of HSPG2 and a variety of important clinical parameters, such as median white blood cell (WBC) count and bone marrow (BM) blasts, were further analyzed. The expression level of HSPG2 was significantly upregulated in AML patients at the time of diagnosis, downregulated after complete remission and then elevated again at relapse. Moreover, HSPG2 expression was associated with median WBC count (P < 0.001), median hemoglobin (P = 0.02), median platelet count (P = 0.001), and BM blasts (P < 0.001) in AML patients. Patients with high HSPG2 expression had both worse overall survival (OS) (P = 0.001) and poorer leukemia-free survival (LFS) (P = 0.047). In the multivariate analysis model, HSPG2 was identified as an independent prognostic biomarker of AML. Taken together, these results indicate that HSPG2 overexpression was associated with poor prognosis in AML patients, and may be a prognostic biomarker and therapeutic target of AML.
Collapse
|
24
|
Lin CC, Yao CY, Hsu YC, Hou HA, Yuan CT, Li YH, Kao CJ, Chuang PH, Chiu YC, Chen Y, Chou WC, Tien HF. Knock-out of Hopx disrupts stemness and quiescence of hematopoietic stem cells in mice. Oncogene 2020; 39:5112-5123. [PMID: 32533098 DOI: 10.1038/s41388-020-1340-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/18/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023]
Abstract
HOPX is a stem cell marker in hair follicles and intestines. It was shown critical for primitive hematopoiesis. We previously showed an association between higher HOPX expression and clinical characteristics related to stemness and quiescence of leukemic cells in acute myeloid leukemia (AML) patients. To further explore its physiologic functions in hematopoietic system, we generated a mouse model with hematopoietic cell-specific knockout of Hopx (Hopx-/-). In young Hopx-/- mice, the hematopoietic stem cells (HSC) showed decreased reconstitution ability after serial transplantation. Further transcriptomic study revealed decreased HSC signatures in long-term HSCs from the Hopx-/- mice. At 18 months of age, half of the Hopx-/- mice developed cytopenia and splenomegaly. Bone marrow (BM) from the sick mice showed myeloid hyperplasia with predominant mature neutrophils, and decreased progenitor cells and lymphocytes. These phenotypes suggested critical functions of Hopx in maintaining HSC quiescence. Transcriptomic study of the Hopx-/- marrow cells showed significant downregulation of the Cxcl12-Cxcr4 axis, which is critical for maintenance of HSC quiescence. We next examined the role of Hopx in AML by using the MN1 overexpression murine leukemia model. Mice transplanted with MN1-overexpressed Hopx-/- BM cells developed AML with more aggressive phenotypes compared with those transplanted with MN1-overexpressed Hopx-wild cells. Hopx-/- MN1-overexpressed leukemia cells showed higher proliferation rate and downregulation of Cxcl12 and Cxcr4. Furthermore, in human AML, BM plasma CXCL12 levels were lower in patients with lower HOPX expression. In conclusion, our study highlights the roles of Hopx in maintenance of quiescence of the hematopoietic stem cells through CXCL12 pathway in vivo and provides implication of this protein in normal and malignant hematopoiesis.
Collapse
Affiliation(s)
- Chien-Chin Lin
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan
| | - Chi-Yuan Yao
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yueh-Chwen Hsu
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsin-An Hou
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chang-Tsu Yuan
- Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan.,Department of Pathology, Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Hung Li
- Department of Animal Science, Chinese Culture University, Taipei, Taiwan
| | - Chein-Jun Kao
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Po-Han Chuang
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Chiao Chiu
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Yidong Chen
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Wen-Chien Chou
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan. .,Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| | - Hwei-Fang Tien
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
25
|
Wang YH, Lin CC, Yao CY, Hsu CL, Hou HA, Tsai CH, Chou WC, Tien HF. A 4-gene leukemic stem cell score can independently predict the prognosis of myelodysplastic syndrome patients. Blood Adv 2020; 4:644-654. [PMID: 32078680 PMCID: PMC7042996 DOI: 10.1182/bloodadvances.2019001185] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/24/2020] [Indexed: 02/07/2023] Open
Abstract
Myelodysplastic syndrome (MDS) comprised a heterogeneous group of diseases. The prognosis of patients varies even in the same risk groups. Searching for novel prognostic markers is warranted. Leukemic stem cells (LSCs) are responsible for chemoresistance and relapse in leukemia. Recently, expressions of 17 genes related to stemness of LSCs were found to be associated with prognosis in acute myeloid leukemia patients. However, the clinical impact of LSC genes expressions in MDS, a disorder arising from hematopoietic stem cells, remains unclear. We analyzed expression profile of the 17 stemness-related genes in primary MDS patients and identified expression of 4 genes (LAPTM4B, NGFRAP1, EMP1, and CPXM1) were significantly correlated with overall survival (OS). We constructed an LSC4 scoring system based on the weighted sums of the expression of 4 genes and explored its clinical implications in MDS patients. Higher LSC4 scores were associated with higher revised International Prognostic Scoring System (IPSS-R) scores, complex cytogenetics, and mutations in RUNX1, ASXL1, and TP53. High-score patients had significantly shorter OS and leukemia-free survival (LFS), which was also confirmed in 2 independent validation cohorts. Subgroup analysis revealed the prognostic significance of LSC4 scores for OS remained valid across IPSS-R lower- and higher-risk groups. Furthermore, higher LSC4 score was an independent adverse risk factor for OS and LFS in multivariate analysis. In summary, LSC4 score can independently predict prognosis in MDS patients irrespective of IPSS-R risks and may be used to guide the treatment of MDS patients, especially lower-risk group in whom usually only supportive treatment is given.
Collapse
Affiliation(s)
- Yu-Hung Wang
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; and
- Division of Hematology, Department of Internal Medicine
| | - Chien-Chin Lin
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; and
- Division of Hematology, Department of Internal Medicine
- Department of Laboratory Medicine, and
| | - Chi-Yuan Yao
- Division of Hematology, Department of Internal Medicine
- Department of Laboratory Medicine, and
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsin-An Hou
- Division of Hematology, Department of Internal Medicine
| | | | - Wen-Chien Chou
- Division of Hematology, Department of Internal Medicine
- Department of Laboratory Medicine, and
| | | |
Collapse
|
26
|
Zhang J, Wang Y, Mo W, Zhang R, Li Y. The clinical and prognostic significance of FOXN3 downregulation in acute myeloid leukaemia. Int J Lab Hematol 2020; 42:270-276. [PMID: 32078244 PMCID: PMC7317382 DOI: 10.1111/ijlh.13162] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 01/05/2020] [Accepted: 01/17/2020] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The expression of forkhead box N3 (FOXN3), also known as checkpoint suppressor 1 (CHES1), is reduced in many types of tumours. However, the clinical significance of FOXN3 and its potential role in acute myeloid leukaemia (AML) remain largely unknown. METHODS A total of 117 de novo AML patients newly diagnosed between December 2015 and January 2018 were included in this study. The expression of FOXN3 and its clinical significance were analysed in these AML patients. RESULTS The expression of FOXN3 was significantly downregulated in AML. In addition, lower FOXN3 expression was associated with older age and higher white blood cell counts. Moreover, a close correlation was observed between lower FOXN3 expression and a lower complete remission (CR) rate and shorter overall survival (OS), which was further analysed by multivariate analysis. CONCLUSION These data suggest that FOXN3 is a novel biomarker in AML and that lower FOXN3 expression predicts poor chemotherapy response and prognosis in AML.
Collapse
Affiliation(s)
- Jinjing Zhang
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yue Wang
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Wenbin Mo
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Rui Zhang
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yan Li
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
27
|
Fu Y, Xu M, Cui Z, Yang Z, Zhang Z, Yin X, Huang X, Zhou M, Wang X, Chen C. Genome-wide identification of FHL1 as a powerful prognostic candidate and potential therapeutic target in acute myeloid leukaemia. EBioMedicine 2020; 52:102664. [PMID: 32062360 PMCID: PMC7021551 DOI: 10.1016/j.ebiom.2020.102664] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 01/16/2020] [Accepted: 01/22/2020] [Indexed: 01/21/2023] Open
Abstract
Background Acute myeloid leukaemia (AML) is a malignant haematological tumour with high heterogeneity and mortality. A reliable prognostic assessment is critical for treatment strategies. However, the current prognostic evaluation system of AML is insufficient. Methods Genome-wide univariate Cox regression analysis was performed on three independent AML datasets to screen for the prognostic-related genes. Kaplan–Meier survival analysis was employed to verify the efficacy of FHL1 in evaluating overall survival in 1298 de novo AML patients, 648 non-acute promyelocytic leukaemia AML patients and 407 cytogenetically normal AML patients; the data for some of these patients were also used for EFS and RFS validation. Multivariate Cox regression was performed to validate FHL1 as an independent prognostic indicator. WGCNA, GSEA, and gene correlation analysis were applied to explore the mechanism of FHL1 in AML. The synergistic cytocidal effect of FHL1 knockdown was verified in in vitro experiments. Findings Comprehensive genome-wide analyses and large-sample validation showed that FHL1 is a powerful prognostic candidate for overall survival, event-free survival, and relapse-free survival in AML and is independent of prognosis-related clinical factors and genetic abnormalities. The molecular mechanism may occur through regulation of FHL1 in leukaemia stem cells, tumour-associated signalling pathways, and transmembrane transport of chemotherapeutic drugs. FHL1-targeted intervention enhances the sensitivity of AML cells to cytarabine. Interpretation FHL1 may serve as an evaluation factor for clinical strategy selection, and its targeted intervention may be beneficial for chemotherapy in AML patients.
Collapse
Affiliation(s)
- Yue Fu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China; School of Medicine, Shandong University, Jinan, Shandong, China; Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Man Xu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zelong Cui
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zongcheng Yang
- School of Stomatology, Shandong University, Jinan, Shandong, China
| | - Zhiyong Zhang
- School of Computer Science and Technology, Shandong University, Qingdao, Shandong, China; Fintech Institute of the People's Bank of China, Shenzhen, Guangdong, China
| | - Xiaolin Yin
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiangnan Huang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Minran Zhou
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaoming Wang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Chunyan Chen
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
28
|
Xu ZJ, Gu Y, Wang CZ, Jin Y, Wen XM, Ma JC, Tang LJ, Mao ZW, Qian J, Lin J. The M2 macrophage marker CD206: a novel prognostic indicator for acute myeloid leukemia. Oncoimmunology 2019; 9:1683347. [PMID: 32002295 PMCID: PMC6959428 DOI: 10.1080/2162402x.2019.1683347] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 01/13/2023] Open
Abstract
Hematological malignancies possess a distinctive immunologic microenvironment compared with solid tumors. Here, using an established computational algorithm (CIBERSORT), we systematically analyzed the overall distribution of 22 tumor-infiltrating leukocyte (TIL) populations in more than 2000 bone marrow (BM) samples from 5 major hematological malignancies and healthy controls. Focusing on significantly altered TILs in acute myeloid leukemia (AML), we found that patients with AML exhibited increased frequencies of M2 macrophages, compared to either healthy controls or the other four malignancies. High infiltration of M2 macrophages was associated with poor outcome in AML. Further analysis revealed that CD206, a M2 marker gene, could faithfully reflect variation in M2 fractions and was more highly expressed in AML than normal controls. High CD206 expression predicted inferior overall survival (OS) and event-free survival (EFS) in two independent AML cohorts. Among 175 patients with intermediate-risk cytogenetics, the survival still differed greatly between low and high CD206 expressers (OS; P < .0001; 3-year rates, 56% v 32%; EFS; P < .001; 3-year rates, 47% v 25%). When analyzed in a meta-analysis, CD206 as a continuous variable showed superior predictive performance than classical prognosticators in AML (BAALC, ERG, EVI1, MN1, and WT1). In summary, M2 macrophages are preferentially enriched in AML. The M2 marker CD206 may serve as a new prognostic marker in AML.
Collapse
Affiliation(s)
- Zi-Jun Xu
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, P.R. China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu, P.R. China
| | - Yu Gu
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, P.R. China.,Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| | - Cui-Zhu Wang
- Department of Oncology, Affiliated Haian Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| | - Ye Jin
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, P.R. China.,Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| | - Xiang-Mei Wen
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, P.R. China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu, P.R. China
| | - Ji-Chun Ma
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, P.R. China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu, P.R. China
| | - Li-Juan Tang
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, P.R. China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu, P.R. China
| | - Zhen-Wei Mao
- The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu, P.R. China
| | - Jun Qian
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, P.R. China.,Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| | - Jiang Lin
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, P.R. China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu, P.R. China
| |
Collapse
|
29
|
Yang L, Shen K, Zhang M, Zhang W, Cai H, Lin L, Long X, Xing S, Tang Y, Xiong J, Wang J, Li D, Zhou J, Xiao M. Clinical Features and MicroRNA Expression Patterns Between AML Patients With DNMT3A R882 and Frameshift Mutations. Front Oncol 2019; 9:1133. [PMID: 31709191 PMCID: PMC6821681 DOI: 10.3389/fonc.2019.01133] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/10/2019] [Indexed: 01/01/2023] Open
Abstract
Background: DNA methyltransferase 3A (DNMT3A) plays a unique role in hematopoiesis and acute myeloid leukemia (AML) pathogenesis. While the influences of DNMT3A mutation subtypes are still under debate. Purpose: Exploration of the clinical and molecular differences between AML patients carrying DNMT3A R882 mutations and DNMT3A frameshift mutations. Methods: Next generation of sequencing (NGS) and clinical data of 118 AML patients in our center were analyzed and compared. NGS, mRNA and miRNA profiling and clinical data from 12 patients in TCGA database were integrative analyzed. Results: Among all patients enrolled, 113 patients were positive for the variants of interest. Overall, a total of 295 variants were discovered, among which 24 DNMT3A mutations were detected, including 1 non-sense, 20 missense, 3 frameshift mutations. And 7 DNMT3A R882 mutations (3 R882H, 2 R882C, and 2 R882P) were found. Clinical analysis from our cohort and TCGA database indicated that patients carrying DNMT3A R882 mutation exhibited significantly higher levels of peripheral blood hemoglobin and non-significantly inferior prognosis compared with patients with DNMT3A frameshift mutations. Integrative analysis indicated that miR-10b, miR-143, and miR-30a were significantly decreased in the DNMT3A R882 group. High miR-143 expression is significantly associated with better prognosis in AML patients with DNMT3A mutations. Conclusion: Different molecular and clinical characteristics existed between patients with DNMT3A variant subtypes. The distinct microRNA expression pattern for DNMT3A R882 AML patients might not only act as markers to predict disease prognosis, but also could be further investigated to develop novel therapeutic targets for patients with DNMT3A mutations.
Collapse
Affiliation(s)
- Li Yang
- Department of Hematology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ke'Feng Shen
- Department of Hematology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Mei'Lan Zhang
- Department of Hematology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Zhang
- Department of Hematology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Hao'Dong Cai
- Department of Hematology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Li'Man Lin
- Department of Hematology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao'Lu Long
- Department of Hematology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Shu'Gang Xing
- Department of Hematology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Tang
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Xiong
- Department of Hematology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jia'Chen Wang
- Department of Hematology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Deng'Ju Li
- Department of Hematology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jian'Feng Zhou
- Department of Hematology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Min Xiao
- Department of Hematology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Lima EU, Rubio IGS, Da Silva JC, Galrão AL, Pêssoa D, Oliveira TC, Carrijo F, Silva Campos I, Fonseca Espinheira L, Sampaio LJ, Lima CR, Cerutti JM, Ramos HE. HOPX homeobox methylation in differentiated thyroid cancer and its clinical relevance. Endocr Connect 2018; 7:1333-1342. [PMID: 30400039 PMCID: PMC6280589 DOI: 10.1530/ec-18-0380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 10/24/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND The inactivation of the tumor-suppressor homeodomain-only protein X (HOPX) usually involves promoter methylation in several cancer types. This study aimed to investigate the HOPX-β mRNA expression and promoter methylation and their clinical relevance in differentiated thyroid cancer (DTC). PATIENTS AND METHODS Clinicopathological data and paraffin-embedded thyroid tumor tissues from 21 patients with DTC and 6 with benign tumors (T) and their non-tumor parenchyma (NT) were investigated. Tumor cell lines (FTC238, FTC236 and WRO) were treated with demethylating agent. HOPX-β mRNA expression was assessed by qRT-PCR and methylation status by Q-MSP. Thyroid cancer data from Cancer Genome Atlas (TCGA) was also collected. RESULTS HOPX-β mRNA re-expression in two cell lines treated with demethylating agent was observed concomitantly with reduced promoter methylation. Reduced mRNA expression in T group compared to their NT was observed, and reduced protein expression in T compared to NT was observed in three cases. Low mRNA expression with high methylation status was detected in 6/14 DTC samples. High methylation status was associated with older age at diagnosis, recurrent or progressive disease and with the presence of new neoplasm event post initial therapy while hyper-methylation correlated with worse overall survival, worse disease-free status and older age. CONCLUSION A moderate coupling of downregulation of HOPX-β mRNA expression in DTC followed by high HOPX-β promoter methylation was observed however; high HOPX promoter methylation status was associated with the worse prognosis of DTC patients.
Collapse
Affiliation(s)
- Erika Urbano Lima
- Biological Science Department, Thyroid Molecular Sciences Laboratory, Universidade Federal de São Paulo, São Paulo, Brazil
- Structural and Functional Biology Program, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ileana G S Rubio
- Biological Science Department, Thyroid Molecular Sciences Laboratory, Universidade Federal de São Paulo, São Paulo, Brazil
- Structural and Functional Biology Program, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Joaquim Custodio Da Silva
- Department of Bio-regulation, Thyroid Study Laboratory, Health & Science Institute, Federal University of Bahia, Salvador, Brazil
- Post-graduate Program in Interactive Processes of Organs and Systems, Health & Science Institute, Federal University of Bahia, Salvador, Brazil
| | - Ana Luiza Galrão
- Biological Science Department, Thyroid Molecular Sciences Laboratory, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Danielle Pêssoa
- Department of Bio-regulation, Thyroid Study Laboratory, Health & Science Institute, Federal University of Bahia, Salvador, Brazil
- Post-graduate Program in Interactive Processes of Organs and Systems, Health & Science Institute, Federal University of Bahia, Salvador, Brazil
| | - Taise Cerqueira Oliveira
- Department of Bio-regulation, Thyroid Study Laboratory, Health & Science Institute, Federal University of Bahia, Salvador, Brazil
| | - Fabiane Carrijo
- Department of Bio-regulation, Thyroid Study Laboratory, Health & Science Institute, Federal University of Bahia, Salvador, Brazil
- Post-graduate Program in Interactive Processes of Organs and Systems, Health & Science Institute, Federal University of Bahia, Salvador, Brazil
| | | | - Luciano Fonseca Espinheira
- Department of Pathology, Sao Rafael Hospital, Salvador, Brazil
- Department of Anatomic Pathology & Legal Medicine, Bahia Federal Medical School, Federal University of Bahia, Salvador, Brazil
| | | | | | - Janete Maria Cerutti
- Structural and Functional Biology Program, Universidade Federal de São Paulo, São Paulo, Brazil
- Division of Genetics, Department of Morphology and Genetics, Genetic Basis of Thyroid Tumors Laboratory, Paulista School of Medicine, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Helton Estrela Ramos
- Department of Bio-regulation, Thyroid Study Laboratory, Health & Science Institute, Federal University of Bahia, Salvador, Brazil
- Post-graduate Program in Interactive Processes of Organs and Systems, Health & Science Institute, Federal University of Bahia, Salvador, Brazil
- Correspondence should be addressed to H E Ramos:
| |
Collapse
|
31
|
Liu B, Li LJ, Gong X, Zhang W, Zhang H, Zhao L. Co-expression of ATP binding cassette transporters is associated with poor prognosis in acute myeloid leukemia. Oncol Lett 2018; 15:6671-6677. [PMID: 29616130 DOI: 10.3892/ol.2018.8095] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 02/13/2018] [Indexed: 01/08/2023] Open
Abstract
Chemotherapy failure remains a challenge when treating patients with acute myeloid leukemia (AML), who often suffer from persistent or relapsed disease. The multidrug resistance (MDR) mediated by efflux transporters of the ATP binding cassette (ABC) superfamily is a major obstacle for successful chemotherapy. The present study aimed to elucidate whether the expression of ABC transporters was associated with prognostic factors and responses to chemotherapy in patients with AML, with particular focus on whether co-expression of multiple ABC transporters resulted in a worse prognosis. In the present study, the mRNA expression levels of ABC transporters ABCB1, ABCB4, ABCC1, ABCC4 and ABCG2 in bone marrow (BM) mononuclear cell (MNC) samples from 96 de novo patients with AML and in the peripheral blood (PB) MNC samples from 22 normal individuals were investigated using reverse transcription-quantitative polymerase chain reaction analysis. It was revealed that ABCB1, ABCC1, ABCC4 and ABCG2 were expressed at higher levels in patients with AML compared with normal individuals, whereas ABCB4 had a lower expression level. The expression of ABCB4 in patients with AML was significantly lower than in normal individuals (P<0.001). Patients risk status was associated with ABCB1 (P=0.037), ABCC1 (P=0.047), ABCC4 (P=0.015) and ABCG2 (P=0.027). The 4 genes were expressed a significantly higher levels in the poor response group compared with the good response group (ABCB1, P=0.014; ABCC1, P=0.021; ABCC4, P=0.005; ABCG2, P=0.009). The overexpression of the 4 ABC transporters and the complete remission rate were inversely correlated (P<0.001). These results suggest that the co-expression of multiple ABC transporters may contribute to a worse prognosis in AML.
Collapse
Affiliation(s)
- Bei Liu
- The First Clinical Medical College of Lanzhou University, The First Affiliated Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China.,Department of Hematology, The First Affiliated Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Li-Jun Li
- The First Clinical Medical College of Lanzhou University, The First Affiliated Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Xia Gong
- Department of Special Medicine, The First Affiliated Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Wei Zhang
- Central Laboratory, The First Affiliated Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Hui Zhang
- The First Clinical Medical College of Lanzhou University, The First Affiliated Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Li Zhao
- The First Clinical Medical College of Lanzhou University, The First Affiliated Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China.,Central Laboratory, The First Affiliated Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
32
|
Nakamura K, Akiba J, Ogasawara S, Naito Y, Nakayama M, Abe Y, Kusukawa J, Yano H. SUOX is negatively associated with multistep carcinogenesis and proliferation in oral squamous cell carcinoma. Med Mol Morphol 2017; 51:102-110. [PMID: 29280012 DOI: 10.1007/s00795-017-0177-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 12/18/2017] [Indexed: 12/15/2022]
Abstract
Oral squamous cell carcinoma (OSCC) is the most common malignant tumor in the head and neck region. The aim of this study was to identify the key molecules and to elucidate the molecular mechanisms of OSCC carcinogenesis through a microarray analysis of RNA extracted from normal epithelium, dysplasia, and squamous cell carcinoma components. Out of molecules that showed changes in gene expression in the microarray analysis, we focused on Sulfite oxidase (SUOX), which correlated significantly with carcinogenic process and exhibited a stepwise decrease in expression. The expression of SUOX was evaluated in detail at the protein level using samples from 58 patients with cancer of the tongue, and correlating clinicopathological factors were also comprehensively examined. SUOX expression declined significantly from normal epithelium to dysplasia to squamous cell carcinoma components in line with carcinogenic process. With regard to squamous cell carcinoma, SUOX expression was significantly lower when T classification was high. Our findings indicated that SUOX is negatively associated with the progression and proliferation of tongue cancer, and suggest that SUOX may be a key molecule in tongue tumors.
Collapse
Affiliation(s)
- Ken Nakamura
- Department of Pathology, Kurume University School of Medicine, 67 Asahimachi, Kurume, 830-0011, Japan
- Dental and Oral Medical Center, Kurume University School of Medicine, Kurume, Japan
| | - Jun Akiba
- Department of Pathology, Kurume University School of Medicine, 67 Asahimachi, Kurume, 830-0011, Japan.
- Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Japan.
| | - Sachiko Ogasawara
- Department of Pathology, Kurume University School of Medicine, 67 Asahimachi, Kurume, 830-0011, Japan
| | - Yoshiki Naito
- Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Japan
| | - Masamichi Nakayama
- Department of Pathology, Kurume University School of Medicine, 67 Asahimachi, Kurume, 830-0011, Japan
| | - Yushi Abe
- Department of Pathology, Kurume University School of Medicine, 67 Asahimachi, Kurume, 830-0011, Japan
- Dental and Oral Medical Center, Kurume University School of Medicine, Kurume, Japan
| | - Jingo Kusukawa
- Dental and Oral Medical Center, Kurume University School of Medicine, Kurume, Japan
| | - Hirohisa Yano
- Department of Pathology, Kurume University School of Medicine, 67 Asahimachi, Kurume, 830-0011, Japan
| |
Collapse
|
33
|
Zhang Q, Dai K, Bi L, Jiang S, Han Y, Yu K, Zhang S. Pretreatment platelet count predicts survival outcome of patients with de novo non-M3 acute myeloid leukemia. PeerJ 2017; 5:e4139. [PMID: 29302387 PMCID: PMC5742276 DOI: 10.7717/peerj.4139] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/15/2017] [Indexed: 12/19/2022] Open
Abstract
Background Pretreatment platelet count has been reported as a potential tool to predict survival outcome in several solid tumors. However, the predictive value of pretreatment platelet count remains obscure in de novo acute myeloid leukemia (AML) excluding acute promyelocytic leukemia (M3). Methods We conducted a retrospective review of 209 patients with de novo non-M3 AML in our institute over a period of 8 years (2007–2015). Receiver operating characteristic (ROC) curve analysis was used to determine the optimal platelet (PLT) cutoff in patients. We analyzed the overall survival (OS) and disease free survival (DFS) using the log-rank test and Cox regression analysis. Results By defining the platelet count 50 × 109/L and 120 × 109/L as two cut-off points, we categorized the patients into three groups: low (<50 × 109/L), medium (50–120 × 109/L) and high (>120 × 109/L). On univariate analysis, patients with medium platelet count had longer OS and DFS than those with low or high platelet count. However, the multivariate analysis showed that only longer DFS was observed in patients with medium platelet count than those with low or high platelet count. Conclusion Our findings indicate that pretreatment platelet count has a predictive value for the prognosis of patients with non-M3 AML.
Collapse
Affiliation(s)
- Qianying Zhang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kanchun Dai
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Laixi Bi
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Songfu Jiang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yixiang Han
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kang Yu
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shenghui Zhang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Division of Clinical Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
34
|
Bell AH, DeMonte F, Raza SM, Rhines LD, Tatsui CE, Prieto VG, Fuller GN, Bell D. Transcriptome comparison identifies potential biomarkers of spine and skull base chordomas. Virchows Arch 2017; 472:489-497. [PMID: 28844110 DOI: 10.1007/s00428-017-2224-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/08/2017] [Accepted: 08/20/2017] [Indexed: 10/19/2022]
Abstract
Chordomas are rare, slowly growing, locally aggressive bone neoplasms that arise from embryonic remnants of the notochord, showing dual epithelial-mesenchymal differentiation. The high plasticity probably is the main reason for the high variety in phenotypes of chordoma, from its high heterogeneity on a cellular level to its subtype variations depending on tissue location, with its potential to develop from an inactive quiescent form to an aggressive cancer with extreme adaptability and resistance to drugs and other treatments. Gene expression profiles of formalin-fixed, paraffin-embedded skull chordoma, spine chordoma, and normal tissue specimens were generated and compared. Using strict criteria, we identified 222 differentially expressed transcripts unique to skull base chordoma, 261 unique to spine chordoma, and 192 common to both chordoma subtypes. Further analysis of these three groups of transcripts allowed the selection of three subsets of highly differentially expressed genes as potential biomarkers, disease drivers, and therapeutic targets in both chordoma subtypes. Immunohistochemistry revealed LMX1A to be dominant in skull base chordoma, SALL3 to be unique to spine chordoma, and T to be common to both chordoma subtypes. In both chordoma subtypes, the genes with the highest expression were predominantly development-related genes, mostly transcription factors. Our findings indicate that these developmental genes play important oncogenic roles in chordoma, mainly causing high plasticity and resistance to therapy in both these cancer subtypes but also determining their differentiation status and proliferation activity, pointing to features expected of heterogeneous stem cell-like tissues with similarities to their notochord origins.
Collapse
Affiliation(s)
- Achim H Bell
- Pathology Department, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.,Pathology Research Department, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Franco DeMonte
- Head and Neck Surgery Department, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Neurosurgery Department, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shaan M Raza
- Head and Neck Surgery Department, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Neurosurgery Department, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Laurence D Rhines
- Neurosurgery Department, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Claudio E Tatsui
- Neurosurgery Department, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Victor G Prieto
- Pathology Department, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Gregory N Fuller
- Pathology Department, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Diana Bell
- Pathology Department, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA. .,Head and Neck Surgery Department, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|