1
|
Li J, Wang Y, Wei S, Xu S, Dai S, Zhang L, Tian Z, Zhao L, Lv H. NEK2 Promotes ESCC Malignant Progression by Inhibiting Cellular Senescence via the FOXM1/c-Myc/p27 Signaling Pathway. Mol Carcinog 2025; 64:244-259. [PMID: 39503194 DOI: 10.1002/mc.23839] [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: 06/21/2024] [Revised: 10/15/2024] [Accepted: 10/20/2024] [Indexed: 01/15/2025]
Abstract
Never in mitosis gene A (NIMA)-related kinase 2 (NEK2) is a crucial serine-threonine kinase involved in the process of cell mitosis. However, the precise relationship between NEK2 and esophageal squamous cell carcinoma (ESCC) remains inadequately understood. NEK2 expression in ESCC tissues was assessed through bioinformatics analysis, reverse transcription-quantitative PCR (RT-qPCR) and immunohistochemistry, revealing a correlation with ESCC patient prognosis. Cultured ESCC cells and human normal esophageal epithelial cells (HEEC) were used to investigate the effects of NEK2 knockdown on the development and progression of ESCC by integrated confluence algorithm, colony formation, wound-healing, transwell, and ESCC xenograft tumor model, in vitro and in vivo. In ESCC tissues, NEK2 was found to be significantly upregulated, and its expression correlated with poor prognosis in ESCC patients. NEK2 may facilitate ESCC development by regulating cell proliferation, migration, and invasion. Additionally, results from in vivo experiments suggested that NEK2 knockdown can inhibit tumor growth. Moreover, forkhead box M1 (FOXM1) was identified as a potential downstream target of NEK2 in the regulation of ESCC, with its overexpression reversing the effects of NEK2 knockdown on ESCC. Mechanistic studies also indicated that NEK2 may promote the malignant progression of ESCC by inhibiting cellular senescence through the activation of the FOXM1/c-Myc/p27 signaling pathways, which may provide a novel perspective for the management of ESCC.
Collapse
Affiliation(s)
- Jiachen Li
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yaojie Wang
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Tumor Gene Diagnosis, Prevention, and Therapy of Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Sisi Wei
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Tumor Gene Diagnosis, Prevention, and Therapy of Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shi Xu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Suli Dai
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Tumor Gene Diagnosis, Prevention, and Therapy of Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Li Zhang
- Department of Geriatric, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ziqiang Tian
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lianmei Zhao
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Tumor Gene Diagnosis, Prevention, and Therapy of Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huilai Lv
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
2
|
Dong Q, Wang D, Song C, Gong C, Liu Y, Zhou X, Yue J, Hu Y, Liu H, Zhu L, Niu X, Zheng T, Zhang X, Jin J, Wang T, Ju R, Wang C, Jiang Q, Gao T, Jin Y, Li P, Wang Y, Zhang C, Wang GF, Cao C, Liu X. ABL1-mediated phosphorylation promotes FOXM1-related tumorigenicity by Increasing FOXM1 stability. Cell Death Differ 2024; 31:1285-1301. [PMID: 39060421 PMCID: PMC11445503 DOI: 10.1038/s41418-024-01339-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 06/20/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
The transcription factor FOXM1, which plays critical roles in cell cycle progression and tumorigenesis, is highly expressed in rapidly proliferating cells and various tumor tissues, and high FOXM1 expression is related to a poor prognosis. However, the mechanism responsible for FOXM1 dysregulation is not fully understood. Here, we show that ABL1, a nonreceptor tyrosine kinase, contributes to the high expression of FOXM1 and FOXM1-dependent tumor development. Mechanistically, ABL1 directly binds FOXM1 and mediates FOXM1 phosphorylation at multiple tyrosine (Y) residues. Among these phospho-Y sites, pY575 is indispensable for FOXM1 stability as phosphorylation at this site protects FOXM1 from ubiquitin-proteasomal degradation. The interaction of FOXM1 with CDH1, a coactivator of the E3 ubiquitin ligase anaphase-promoting complex/cyclosome (APC/C), which is responsible for FOXM1 degradation, is significantly inhibited by Y575 phosphorylation. The phospho-deficient FOXM1(Y575F) mutant exhibited increased ubiquitination, a shortened half-life, and consequently a substantially decreased abundance. Compared to wild-type cells, a homozygous Cr-Y575F cell line expressing endogenous FOXM1(Y575F) that was generated by CRISPR/Cas9 showed obviously delayed mitosis progression, impeded colony formation and inhibited xenotransplanted tumor growth. Overall, our study demonstrates that ABL1 kinase is involved in high FOXM1 expression, providing clear evidence that ABL1 may act as a therapeutic target for the treatment of tumors with high FOXM1 expression.
Collapse
Affiliation(s)
- Qincai Dong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Di Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Caiwei Song
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Chunxue Gong
- Institute of Health Sciences, Anhui University, Hefei, 230601, China
| | - Yue Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Xinwei Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Junjie Yue
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Yong Hu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Hainan Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Lin Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Xiayang Niu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Tong Zheng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Xun Zhang
- Institute of Health Sciences, Anhui University, Hefei, 230601, China
| | - Jing Jin
- Institute of Health Sciences, Anhui University, Hefei, 230601, China
| | - Tingting Wang
- Institute of Health Sciences, Anhui University, Hefei, 230601, China
| | - Ruixia Ju
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Chen Wang
- Institute of Health Sciences, Anhui University, Hefei, 230601, China
| | - Qian Jiang
- Institute of Health Sciences, Anhui University, Hefei, 230601, China
| | - Ting Gao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Yanwen Jin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Ping Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Yan Wang
- Clinical Biobank Center, Medical Innovation Research Division, Chinese PLA General Hospital, 100853, Beijing, China
| | - Chunmei Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Guang-Fei Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China.
| | - Cheng Cao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China.
| | - Xuan Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China.
| |
Collapse
|
3
|
He L, Qian H, Seyiti A, Yang C, Shi N, Chen C, Zhang P, Hou Y. CD133 +/ABCC5 + cervical cancer cells exhibit cancer stem cell properties. Heliyon 2024; 10:e37066. [PMID: 39296204 PMCID: PMC11408061 DOI: 10.1016/j.heliyon.2024.e37066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/21/2024] Open
Abstract
Objective This study explores the correlation between Forkhead box M1 (FOXM1) and ATP-binding cassette subfamily C member 5 (ABCC5) in relation to paclitaxel resistance in cervical cancer. It aims to identify potential cervical cancer stem cell markers, offering fresh perspectives for developing therapeutic strategies to overcome paclitaxel chemoresistance in cervical cancer. Methods Paclitaxel-resistant Hela cells (Hela/Taxol) were developed by intermittently exposing Hela cells to progressively increasing concentrations of paclitaxel. We assessed the biological properties of both Hela and Hela/Taxol cells using various assays: cell proliferation, clonogenic, cell cycle, apoptosis, scratch, and transwell. To determine which markers better represent tumor stem cells, we analyzed various known and potential stem cell markers in combination. Flow cytometry was employed to measure the proportion of positive markers in both parental and drug-resistant cell lines. Following statistical analysis to establish relative stability, CD133+ABCC5+ cells were sorted for further examination. Subsequent tests included sphere-forming assays and Western blot analysis to detect the presence of the stem cell-specific protein Sox2, aiding in the identification of viable cervical cancer stem cell markers. Results The Hela/Taxol cell line exhibited significantly enhanced proliferation, migration, and invasion capabilities compared to the Hela cell line, alongside a marked reduction in apoptosis rates (P < 0.01). Notably, proportions of CD44+, CD24+CD44+, ABCC5+, CD24+CD44+ABCC5+, CD44+ABCC5+, CD24+CD44+FOXM1+, CD44+FOXM1+, CD133+ABCC5+, and CD133+FOXM1+ were significantly higher (P < 0.05). Furthermore, the size and number of spheres formed byCD133+ABCC5+ cells were greater in the sorted Hela/Taxol line (P < 0.01), with increased expression of the stem cell marker Sox2 (P < 0.001). Conclusion The Hela/Taxol cells demonstrate increased tumoral stemness, suggesting that CD133+ABCC5+ may serve as a novel marker for cervical cancer stem cells.
Collapse
Affiliation(s)
- Lin He
- Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830011, PR China
| | - Hengjun Qian
- Yibin Second People's Hospital, Sichuan, 644002, PR China
| | - Ayinuer Seyiti
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang, 830011, PR China
| | - Chengshaoxiong Yang
- Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830011, PR China
| | - Ning Shi
- Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830011, PR China
| | - Chen Chen
- Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830011, PR China
| | - Pingxu Zhang
- Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830011, PR China
| | - Youxiang Hou
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang, 830011, PR China
| |
Collapse
|
4
|
Xian M, Wang Q, Xiao L, Zhong L, Xiong W, Ye L, Su P, Zhang C, Li Y, Orlowski RZ, Zhan F, Ganguly S, Zu Y, Qian J, Yi Q. Leukocyte immunoglobulin-like receptor B1 (LILRB1) protects human multiple myeloma cells from ferroptosis by maintaining cholesterol homeostasis. Nat Commun 2024; 15:5767. [PMID: 38982045 PMCID: PMC11233649 DOI: 10.1038/s41467-024-50073-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 06/27/2024] [Indexed: 07/11/2024] Open
Abstract
Multiple myeloma (MM) is a hematologic malignancy characterized by uncontrolled proliferation of plasma cells in the bone marrow. MM patients with aggressive progression have poor survival, emphasizing the urgent need for identifying new therapeutic targets. Here, we show that the leukocyte immunoglobulin-like receptor B1 (LILRB1), a transmembrane receptor conducting negative immune response, is a top-ranked gene associated with poor prognosis in MM patients. LILRB1 deficiency inhibits MM progression in vivo by enhancing the ferroptosis of MM cells. Mechanistic studies reveal that LILRB1 forms a complex with the low-density lipoprotein receptor (LDLR) and LDLR adapter protein 1 (LDLRAP1) to facilitate LDL/cholesterol uptake. Loss of LILRB1 impairs cholesterol uptake but activates the de novo cholesterol synthesis pathway to maintain cellular cholesterol homeostasis, leading to the decrease of anti-ferroptotic metabolite squalene. Our study uncovers the function of LILRB1 in regulating cholesterol metabolism and protecting MM cells from ferroptosis, implicating LILRB1 as a promising therapeutic target for MM patients.
Collapse
Affiliation(s)
- Miao Xian
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Qiang Wang
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Liuling Xiao
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Ling Zhong
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Wei Xiong
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Lingqun Ye
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Pan Su
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Chuanchao Zhang
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Yabo Li
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Robert Z Orlowski
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Fenghuang Zhan
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Siddhartha Ganguly
- Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, USA
| | - Youli Zu
- Department of Pathology and Genomic Medicine, Institute for Academic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Jianfei Qian
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Qing Yi
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA.
| |
Collapse
|
5
|
Wu H, Qian J, Zhou L, Hu T, Zhang Y, Wang C, Yang Y, Gu C. FHND004 inhibits malignant proliferation of multiple myeloma by targeting PDZ-binding kinase in MAPK pathway. Aging (Albany NY) 2024; 16:4811-4831. [PMID: 38460944 PMCID: PMC10968680 DOI: 10.18632/aging.205634] [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: 11/01/2023] [Accepted: 02/08/2024] [Indexed: 03/11/2024]
Abstract
Inhibitors of Epidermal growth factor receptor tyrosine kinase (EGFR-TKIs) are producing impressive benefits to responsive types of cancers but challenged with drug resistances. FHND drugs are newly modified small molecule inhibitors based on the third-generation EGFR-TKI AZD9291 (Osimertinib) that are mainly for targeting the mutant-selective EGFR, particularly for the non-small cell lung cancer (NSCLC). Successful applications of EGFR-TKIs to other cancers are less certain, thus the present pre-clinical study aims to explore the anticancer effect and downstream targets of FHND in multiple myeloma (MM), which is an incurable hematological malignancy and reported to be insensitive to first/second generation EGFR-TKIs (Gefitinib/Afatinib). Cell-based assays revealed that FHND004 and FHND008 significantly inhibited MM cell proliferation and promoted apoptosis. The RNA-seq identified the involvement of the MAPK signaling pathway. The protein chip screened PDZ-binding kinase (PBK) as a potential drug target. The interaction between PBK and FHND004 was verified by molecular docking and microscale thermophoresis (MST) assay with site mutation (N124/D125). Moreover, the public clinical datasets showed high expression of PBK was associated with poor clinical outcomes. PBK overexpression evidently promoted the proliferation of two MM cell lines, whereas the FHND004 treatment significantly inhibited survival of 5TMM3VT cell-derived model mice and growth of patient-derived xenograft (PDX) tumors. The mechanistic study showed that FHND004 downregulated PBK expression, thus mediating ERK1/2 phosphorylation in the MAPK pathway. Our study not only demonstrates PBK as a promising novel target of FHND004 to inhibit MM cell proliferation, but also expands the EGFR kinase-independent direction for developing anti-myeloma therapy.
Collapse
Affiliation(s)
- Hongjie Wu
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinjun Qian
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lianxin Zhou
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tingting Hu
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuanjiao Zhang
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chen Wang
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ye Yang
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chunyan Gu
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
6
|
Mehdi SJ, Ghatak K, Ling W, Johnson SK, Epstein J, Nookaew I, Zangari M, Schinke C, Thanendrarajan S, van Rhee F, Yaccoby S. Growth and dormancy control of myeloma cells by mesenchymal stem cells. Leuk Res 2023; 133:107355. [PMID: 37499483 DOI: 10.1016/j.leukres.2023.107355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/29/2023]
Abstract
Bone marrow mesenchymal stem cells (MSCs) may have contrasting impacts on the progression of multiple myeloma (MM). Priming normal MSCs, by culturing them with MM cells, mimics the MSC-induced MM growth. We studied the contrasting effects of conditioned medium (CM) from unprimed or primed MSCs on growth of MM cells from newly diagnosed cases. We elucidated potential molecular pathways using global gene expression profiling and focused on the role of the mTOR2 component, RICTOR, as a novel mediator of dormancy in MM. Primed MSCs CM consistently increased proportions of proliferating cells and supported MM growth in 3-day (n = 20) and 10-day (n = 12) cultures, effects that were partially mediated through the IGF1 axis. In contrast, unprimed MSCs CM inhibited growth of MM cells in cases mainly from stages I/II MM. The genes most overexpressed in MM cells treated with primed MSCs CM were associated with cell cycle, DNA-damage repair, and proliferation; genes most overexpressed in MM cells treated with unprimed MSCs CM were associated with dormancy pathways including RICTOR (mTOR2 pathway), CXCR4, and BCL2. RICTOR protein level was induced by unprimed MSCs CM and was lower in KI67+ proliferating MM cells treated with primed MSCs CM. RICTOR was underexpressed in clinical relapse samples compared with baseline samples of the same patients. Inhibiting RICTOR expression in primary MM cells promoted their growth, and enforced expression of RICTOR in MM cell lines inhibited their growth. Our findings suggest that, after prolonged interactions with MM cells, bone marrow MSCs shift from MM-repressive to MM-permissive. AVAILABILITY OF DATA AND MATERIALS: Our institutional GEP data of MM cells from newly diagnosed patients used to show RICTOR expression have been deposited at Gene Expression Omnibus (GEO: GSE2658, https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE2658).
Collapse
Affiliation(s)
- Syed J Mehdi
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Kalyan Ghatak
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Wen Ling
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Sarah K Johnson
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Joshua Epstein
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Intawat Nookaew
- Department of Biomedical Informatics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Maurizio Zangari
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Carolina Schinke
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Sharmilan Thanendrarajan
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Frits van Rhee
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Shmuel Yaccoby
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
7
|
Wei R, Zhu Y, Zhang Y, Zhao W, Yu X, Wang L, Gu C, Gu X, Yang Y. AIMP1 promotes multiple myeloma malignancy through interacting with ANP32A to mediate histone H3 acetylation. CANCER COMMUNICATIONS (LONDON, ENGLAND) 2022; 42:1185-1206. [PMID: 36042007 DOI: 10.1002/cac2.12356] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 05/23/2022] [Accepted: 08/16/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Multiple myeloma (MM) is the second most common hematological malignancy. An overwhelming majority of patients with MM progress to serious osteolytic bone disease. Aminoacyl-tRNA synthetase-interacting multifunctional protein 1 (AIMP1) participates in several steps during cancer development and osteoclast differentiation. This study aimed to explore its role in MM. METHODS The gene expression profiling cohorts of MM were applied to determine the expression of AIMP1 and its association with MM patient prognosis. Enzyme-linked immunosorbent assay, immunohistochemistry, and Western blotting were used to detect AIMP1 expression. Protein chip analysis, RNA-sequencing, and chromatin immunoprecipitation and next-generation sequencing were employed to screen the interacting proteins and key downstream targets of AIMP1. The impact of AIMP1 on cellular proliferation was determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay in vitro and a xenograft model in vivo. Bone lesions were evaluated using tartrate-resistant acid phosphatase staining in vitro. A NOD/SCID-TIBIA mouse model was used to evaluate the effect of siAIMP1-loaded exosomes on bone lesion formation in vivo. RESULTS AIMP1 expression was increased in MM patients and strongly associated with unfavorable outcomes. Increased AIMP1 expression promoted MM cell proliferation in vitro and in vivo via activation of the mitogen-activated protein kinase (MAPK) signaling pathway. Protein chip assays and subsequent experiments revealed that AIMP1 interacted with acidic leucine-rich nuclear phosphoprotein 32 family member A (ANP32A) to regulate histone H3 acetylation. In addition, AIMP1 increased histone H3 acetylation enrichment function of GRB2-associated and regulator of MAPK protein 2 (GAREM2) to increase the phosphorylation of extracellular-regulated kinase 1/2 (p-ERK1/2). Furthermore, AIMP1 promoted osteoclast differentiation by activating nuclear factor of activated T cells c1 (NFATc1) in vitro. In contrast, exosome-coated small interfering RNA of AIMP1 effectively suppressed MM progression and osteoclast differentiation in vitro and in vivo. CONCLUSIONS Our data demonstrate that AIMP1 is a novel regulator of histone H3 acetylation interacting with ANP32A in MM, which accelerates MM malignancy via activation of the MAPK signaling pathway.
Collapse
Affiliation(s)
- Rongfang Wei
- Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210001, P. R. China.,School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, P. R. China
| | - Yan Zhu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, P. R. China
| | - Yuanjiao Zhang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, P. R. China
| | - Wene Zhao
- Department of Analytical and Testing Center, Nanjing Medical University, Nanjing, Jiangsu, 211112, P. R. China
| | - Xichao Yu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, P. R. China
| | - Ling Wang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, P. R. China
| | - Chunyan Gu
- Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210001, P. R. China.,School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, P. R. China
| | - Xiaosong Gu
- Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210001, P. R. China.,School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, P. R. China.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, Jiangsu, 226019, P. R. China
| | - Ye Yang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, P. R. China
| |
Collapse
|
8
|
Cheng Y, Sun F, Thornton K, Jing X, Dong J, Yun G, Pisano M, Zhan F, Kim SH, Katzenellenbogen JA, Katzenellenbogen BS, Hari P, Janz S. FOXM1 regulates glycolysis and energy production in multiple myeloma. Oncogene 2022; 41:3899-3911. [PMID: 35794249 PMCID: PMC9355869 DOI: 10.1038/s41388-022-02398-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 06/16/2022] [Accepted: 06/21/2022] [Indexed: 12/16/2022]
Abstract
AbstractThe transcription factor, forkhead box M1 (FOXM1), has been implicated in the natural history and outcome of newly diagnosed high-risk myeloma (HRMM) and relapsed/refractory myeloma (RRMM), but the mechanism with which FOXM1 promotes the growth of neoplastic plasma cells is poorly understood. Here we show that FOXM1 is a positive regulator of myeloma metabolism that greatly impacts the bioenergetic pathways of glycolysis and oxidative phosphorylation (OxPhos). Using FOXM1-deficient myeloma cells as principal experimental model system, we find that FOXM1 increases glucose uptake, lactate output, and oxygen consumption in myeloma. We demonstrate that the novel 1,1-diarylethylene small-compound FOXM1 inhibitor, NB73, suppresses myeloma in cell culture and human-in-mouse xenografts using a mechanism that includes enhanced proteasomal FOXM1 degradation. Consistent with the FOXM1-stabilizing chaperone function of heat shock protein 90 (HSP90), the HSP90 inhibitor, geldanamycin, collaborates with NB73 in slowing down myeloma. These findings define FOXM1 as a key driver of myeloma metabolism and underscore the feasibility of targeting FOXM1 for new approaches to myeloma therapy and prevention.
Collapse
|
9
|
Gu C, Wang Y, Zhang L, Qiao L, Sun S, Shao M, Tang X, Ding P, Tang C, Cao Y, Zhou Y, Guo M, Wei R, Li N, Xiao Y, Duan J, Yang Y. AHSA1 is a promising therapeutic target for cellular proliferation and proteasome inhibitor resistance in multiple myeloma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:11. [PMID: 34991674 PMCID: PMC8734095 DOI: 10.1186/s13046-021-02220-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 12/13/2021] [Indexed: 01/14/2023]
Abstract
BACKGROUND Currently, multiple myeloma (MM) is still an incurable plasma cell malignancy in urgent need of novel therapeutic targets and drugs. METHODS Bufalin was known as a highly toxic but effective anti-cancer compound. We used Bufalin as a probe to screen its potential targets by proteome microarray, in which AHSA1 was the unique target of Bufalin. The effects of AHSA1 on cellular proliferation and drug resistance were determined by MTT, western blot, flow cytometry, immunohistochemistry staining and xenograft model in vivo. The potential mechanisms of Bufalin and KU-177 in AHSA1/HSP90 were verified by co-immunoprecipitation, mass spectrometry, site mutation and microscale thermophoresis assay. RESULTS AHSA1 expression was increased in MM samples compared to normal controls, which was significantly associated with MM relapse and poor outcomes. Furthermore, AHSA1 promoted MM cell proliferation and proteasome inhibitor (PI) resistance in vitro and in vivo. Mechanism exploration indicated that AHSA1 acted as a co-chaperone of HSP90A to activate CDK6 and PSMD2, which were key regulators of MM proliferation and PI resistance respectively. Additionally, we identified AHSA1-K137 as the specific binding site of Bufalin on AHSA1, mutation of which decreased the interaction of AHSA1 with HSP90A and suppressed the function of AHSA1 on mediating CDK6 and PSMD2. Intriguingly, we discovered KU-177, an AHSA1 selective inhibitor, and found KU-177 targeting the same site as Bufalin. Bufalin and KU-177 treatments hampered the proliferation of flow MRD-positive cells in both primary MM and recurrent MM patient samples. Moreover, KU-177 abrogated the cellular proliferation and PI resistance induced by elevated AHSA1, and decreased the expression of CDK6 and PSMD2. CONCLUSIONS We demonstrate that AHSA1 may serve as a promising therapeutic target for cellular proliferation and proteasome inhibitor resistance in multiple myeloma.
Collapse
Affiliation(s)
- Chunyan Gu
- Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yajun Wang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lulin Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Li Qiao
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shanliang Sun
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Miaomiao Shao
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiaozhu Tang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Pinggang Ding
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chao Tang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yuhao Cao
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yanyan Zhou
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Mengjie Guo
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Rongfang Wei
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Nianguang Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yibei Xiao
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Jinao Duan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China. .,State Administration of Traditional Chinese Medicine Key Laboratory of Chinese Medicinal Resources Recycling Utilization, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Ye Yang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
10
|
NAT10 promotes cell proliferation by acetylating CEP170 mRNA to enhance translation efficiency in multiple myeloma. Acta Pharm Sin B 2022; 12:3313-3325. [PMID: 35967285 PMCID: PMC9366180 DOI: 10.1016/j.apsb.2022.01.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/01/2021] [Accepted: 12/16/2021] [Indexed: 12/16/2022] Open
|
11
|
Kuttikrishnan S, Prabhu KS, Khan AQ, Alali FQ, Ahmad A, Uddin S. Thiostrepton inhibits growth and induces apoptosis by targeting FoxM1/SKP2/MTH1 axis in B-precursor acute lymphoblastic leukemia cells. Leuk Lymphoma 2021; 62:3170-3180. [PMID: 34369229 DOI: 10.1080/10428194.2021.1957873] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/19/2021] [Accepted: 07/10/2021] [Indexed: 12/15/2022]
Abstract
Forkhead box M1 (FoxM1) is a transcription factor that plays an important role in the etiology of many cancers, however, its role has not been elucidated in B-precursor acute lymphoblastic leukemia (B-pre-ALL). In the current study, we showed that the downregulation of FoxM1 by its inhibitor thiostrepton inhibited cell viability and induced caspase-dependent apoptosis in a panel of B-pre-ALL cell lines. Thiostrepton led downregulation of FoxM1 accompanied by decreased expression of Aurora kinase A, B, matrix metalloproteinases, and oncogene SKP2 as well as MTH1. Downregulation of the FoxM1/SKP2/MTH1 axis led to increase in the Bax/Bcl2 ratio and suppression of antiapoptotic proteins. Thiostrepton-mediated apoptosis was prevented by N-acetyl cysteine, a scavenger of reactive oxygen species. Co-treatment of B-pre-ALL with subtoxic doses of thiostrepton and bortezomib potentiated the proapoptotic action. Altogether, our results suggest that targeting FoxM1expression could be an attractive strategy for the treatment of B-pre-ALL.
Collapse
Affiliation(s)
- Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- College of Pharmacy, Qatar University, Doha, Qatar
| | - Kirti S Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Abdul Q Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Feras Q Alali
- College of Pharmacy, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University
| | - Aamir Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Laboratory of Animal Research Center, Qatar University, Doha, Qatar
| |
Collapse
|
12
|
Yang B, Diao H, Wang P, Guan F, Liu H. microRNA-877-5p exerts tumor-suppressive functions in prostate cancer through repressing transcription of forkhead box M1. Bioengineered 2021; 12:9094-9102. [PMID: 34654353 PMCID: PMC8806950 DOI: 10.1080/21655979.2021.1989969] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The study aimed to investigate the significant potential role of miR-877-5p in Prostate cancer. The expression levels of miR-877-5p and forkhead box M1 (FOXM1) mRNA were detected by qRT-PCR. The prognostic significance of miR-877-5p in prostate cancer was investigated using Kaplan Meier analysis. Then, Cell Counting Kit-8 (CCK-8) and transwell assay were used to evaluate the effects of miR-877-5p on cell biological functions. The mechanism of miR-877-5p action on prostate cancer cells was investigated by luciferase activity assay with wide-type or mutation. miR-877-5p was lowly expressed both in prostate cancer tissues and cell lines compared with corresponding normal counterparts. Further, miR-877-5p was significantly correlated with Gleason score and TNM stage. Moreover, miR-877-5p may serve as an independent prognostic predictor. In addition, FOXM1 was checked as a direct target gene of miR-877-5p, and miR-877-5p can inhibit the expression of FOXM1 to restrain the growth, migration, and invasion abilities of prostate cancer cells. Taken together, miR-877-5p may act as a suppressor in prostate cancer and reduces cancer cell proliferation, migration and invasion by targeting FOXM1. miR-877-5p may serve as the effective biomarkers and therapeutic target for treating prostate cancer patients.
Collapse
Affiliation(s)
- Bin Yang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Huifeng Diao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Pu Wang
- Department of Urology, Heze Municipal Hospital, Heze, China
| | - Fengju Guan
- Department of Operating Room, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hechen Liu
- Department of Urology, Shandong Provincial Third Hospital, Shandong University, Jinan, China
| |
Collapse
|
13
|
Genetic program activity delineates risk, relapse, and therapy responsiveness in multiple myeloma. NPJ Precis Oncol 2021; 5:60. [PMID: 34183722 PMCID: PMC8239045 DOI: 10.1038/s41698-021-00185-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 05/13/2021] [Indexed: 01/19/2023] Open
Abstract
Despite recent advancements in the treatment of multiple myeloma (MM), nearly all patients ultimately relapse and many become refractory to multiple lines of therapies. Therefore, we not only need the ability to predict which patients are at high risk for disease progression but also a means to understand the mechanisms underlying their risk. Here, we report a transcriptional regulatory network (TRN) for MM inferred from cross-sectional multi-omics data from 881 patients that predicts how 124 chromosomal abnormalities and somatic mutations causally perturb 392 transcription regulators of 8549 genes to manifest in distinct clinical phenotypes and outcomes. We identified 141 genetic programs whose activity profiles stratify patients into 25 distinct transcriptional states and proved to be more predictive of outcomes than did mutations. The coherence of these programs and accuracy of our network-based risk prediction was validated in two independent datasets. We observed subtype-specific vulnerabilities to interventions with existing drugs and revealed plausible mechanisms for relapse, including the establishment of an immunosuppressive microenvironment. Investigation of the t(4;14) clinical subtype using the TRN revealed that 16% of these patients exhibit an extreme-risk combination of genetic programs (median progression-free survival of 5 months) that create a distinct phenotype with targetable genes and pathways.
Collapse
|
14
|
Guo M, Ding P, Zhu Z, Fan L, Zhou Y, Yang S, Yang Y, Gu C. Targeting RFWD2 as an Effective Strategy to Inhibit Cellular Proliferation and Overcome Drug Resistance to Proteasome Inhibitor in Multiple Myeloma. Front Cell Dev Biol 2021; 9:675939. [PMID: 33968945 PMCID: PMC8097052 DOI: 10.3389/fcell.2021.675939] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 03/30/2021] [Indexed: 12/15/2022] Open
Abstract
The potential to overcome resistance to proteasome inhibitors is greatly related with ubiquitin-proteasome system during multiple myeloma (MM) treatment process. The constitutive photomorphogenic 1 (RFWD2), referred to an E3 ubiquitin ligase, has been identified as an oncogene in multiple cancers, yet important questions on the role of RFWD2 in MM biology and treatment remain unclear. Here we demonstrated that MM patients with elevated RFWD2 expression achieved adverse outcome and drug resistance by analyzing gene expression profiling. Moreover, we proved that RFWD2 participated in the process of cell cycle, cell growth and death in MM by mass spectrometry analysis. In vitro study indicated that inducible knockdown of RFWD2 hindered cellular growth and triggered apoptosis in MM cells. Mechanism study revealed that RFWD2 controlled MM cellular proliferation via regulating the degradation of P27 rather than P53. Further exploration unveiled that RFWD2 meditated P27 ubiquitination via interacting with RCHY1, which served as an E3 ubiquitin ligase of P27. Finally, in vivo study illustrated that blocking RFWD2 in BTZ-resistant MM cells overcame the drug resistance in a myeloma xenograft mouse model. Taken together, these findings provide compelling evidence for prompting that targeting RFWD2 may be an effective strategy to inhibit cellular proliferation and overcome drug resistance to proteasome inhibitor in MM.
Collapse
Affiliation(s)
- Mengjie Guo
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Large Data Center, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Pinggang Ding
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhen Zhu
- College of Health and Rehabilitation & College of Acupuncture and Massage, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lu Fan
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yanyan Zhou
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shu Yang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ye Yang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chunyan Gu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Large Data Center, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
15
|
Overexpression of aberrant Wnt5a and its effect on acquisition of malignant phenotypes in adult T-cell leukemia/lymphoma (ATL) cells. Sci Rep 2021; 11:4114. [PMID: 33603066 PMCID: PMC7892546 DOI: 10.1038/s41598-021-83613-2] [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: 08/04/2020] [Accepted: 02/03/2021] [Indexed: 01/31/2023] Open
Abstract
Wnt5a is a ligand of the non-canonical Wnt signaling pathway involved in cell differentiation, motility, and inflammatory response. Adult T-cell leukemia/lymphoma (ATL) is one of the most aggressive T-cell malignancies caused by infection of human T-cell leukemia virus type1 (HTLV-1). Among subtypes of ATL, acute-type ATL cells are particularly resistant to current multidrug chemotherapies and show remarkably high cell-proliferative and invasive phenotypes. Here we show a dramatic increase of WNT5A gene expression in acute-type ATL cells compared with those of indolent-type ATL cells. Treatment with IWP-2 or Wnt5a-specific knockdown significantly suppressed cell growth of ATL-derived T-cell lines. We demonstrated that the overexpression of c-Myb and FoxM1 was responsible for the synergistic activation of the WNT5A promoter. Also, a WNT5A transcript variant without the exon4 (the ΔE4-WNT5A mRNA), encoding ΔC-Wnt5 (1-136aa of 380aa), is overexpressed in acute-type ATL cells. The ΔC-Wnt5a is secreted extracellularly and enhances cellular migration/invasion to a greater extent compared with wildtype (WT)-Wnt5a. Moreover, the ΔC-Wnt5a secretion was not suppressed by IWP-2, indicating that this mutant Wnt5a is secreted via a different pathway from the WT-Wnt5a. Taken together, synergistic overexpression of the ΔC-Wnt5a by c-Myb and FoxM1 may be responsible for the malignant phenotype of acute-type ATL cells.
Collapse
|
16
|
Rocha D, García IA, González Montoro A, Llera A, Prato L, Girotti MR, Soria G, Fernández EA. Pan-Cancer Molecular Patterns and Biological Implications Associated with a Tumor-Specific Molecular Signature. Cells 2020; 10:E45. [PMID: 33396205 PMCID: PMC7823585 DOI: 10.3390/cells10010045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 11/25/2020] [Accepted: 11/26/2020] [Indexed: 12/13/2022] Open
Abstract
Studying tissue-independent components of cancer and defining pan-cancer subtypes could be addressed using tissue-specific molecular signatures if classification errors are controlled. Since PAM50 is a well-known, United States Food and Drug Administration (FDA)-approved and commercially available breast cancer signature, we applied it with uncertainty assessment to classify tumor samples from over 33 cancer types, discarded unassigned samples, and studied the emerging tumor-agnostic molecular patterns. The percentage of unassigned samples ranged between 55.5% and 86.9% in non-breast tissues, and gene set analysis suggested that the remaining samples could be grouped into two classes (named C1 and C2) regardless of the tissue. The C2 class was more dedifferentiated, more proliferative, with higher centrosome amplification, and potentially more TP53 and RB1 mutations. We identified 28 gene sets and 95 genes mainly associated with cell-cycle progression, cell-cycle checkpoints, and DNA damage that were consistently exacerbated in the C2 class. In some cancer types, the C1/C2 classification was associated with survival and drug sensitivity, and modulated the prognostic meaning of the immune infiltrate. Our results suggest that PAM50 could be repurposed for a pan-cancer context when paired with uncertainty assessment, resulting in two classes with molecular, biological, and clinical implications.
Collapse
Affiliation(s)
- Darío Rocha
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina; (D.R.); (A.G.M.)
| | - Iris A. García
- Centro de Investigación y Desarrollo en Inmunología y Enfermedades Infecciosas, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Católica de Córdoba, Córdoba X5016DHK, Argentina;
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina;
| | - Aldana González Montoro
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina; (D.R.); (A.G.M.)
- Facultad de Matemática, Astronomía y Física, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina
| | - Andrea Llera
- Laboratorio de Terapia Molecular y Celular—Genocan, Fundación Instituto Leloir, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires C1405BWE, Argentina;
| | - Laura Prato
- Instituto Académico Pedagógico de Ciencias Básicas y Aplicadas, Universidad Nacional de Villa María, Villa María, Córdoba X5900, Argentina;
| | - María R. Girotti
- Laboratorio de Inmuno Oncología Traslacional, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires C1428ADN, Argentina;
| | - Gastón Soria
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina;
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas, Córdoba X5000HUA, Argentina
| | - Elmer A. Fernández
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina; (D.R.); (A.G.M.)
- Centro de Investigación y Desarrollo en Inmunología y Enfermedades Infecciosas, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Católica de Córdoba, Córdoba X5016DHK, Argentina;
| |
Collapse
|
17
|
Li Z, Yu DS, Doetsch PW, Werner E. Replication stress and FOXM1 drive radiation induced genomic instability and cell transformation. PLoS One 2020; 15:e0235998. [PMID: 33253193 PMCID: PMC7703902 DOI: 10.1371/journal.pone.0235998] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 11/07/2020] [Indexed: 12/25/2022] Open
Abstract
In contrast to the vast majority of research that has focused on the immediate effects of ionizing radiation, this work concentrates on the molecular mechanism driving delayed effects that emerge in the progeny of the exposed cells. We employed functional protein arrays to identify molecular changes induced in a human bronchial epithelial cell line (HBEC3-KT) and osteosarcoma cell line (U2OS) and evaluated their impact on outcomes associated with radiation induced genomic instability (RIGI) at day 5 and 7 post-exposure to a 2Gy X-ray dose, which revealed replication stress in the context of increased FOXM1b expression. Irradiated cells had reduced DNA replication rate detected by the DNA fiber assay and increased DNA resection detected by RPA foci and phosphorylation. Irradiated cells increased utilization of homologous recombination-dependent repair detected by a gene conversion assay and DNA damage at mitosis reflected by RPA positive chromosomal bridges, micronuclei formation and 53BP1 positive bodies in G1, all known outcomes of replication stress. Interference with the function of FOXM1, a transcription factor widely expressed in cancer, employing an aptamer, decreased radiation-induced micronuclei formation and cell transformation while plasmid-driven overexpression of FOXM1b was sufficient to induce replication stress, micronuclei formation and cell transformation.
Collapse
Affiliation(s)
- Zhentian Li
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - David S. Yu
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Paul W. Doetsch
- Laboratory of Genomic Integrity and Structural Biology, NIH, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
| | - Erica Werner
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
18
|
Ordoñez R, Kulis M, Russiñol N, Chapaprieta V, Carrasco-Leon A, García-Torre B, Charalampopoulou S, Clot G, Beekman R, Meydan C, Duran-Ferrer M, Verdaguer-Dot N, Vilarrasa-Blasi R, Soler-Vila P, Garate L, Miranda E, San José-Enériz E, Rodriguez-Madoz JR, Ezponda T, Martínez-Turrilas R, Vilas-Zornoza A, Lara-Astiaso D, Dupéré-Richer D, Martens JHA, El-Omri H, Taha RY, Calasanz MJ, Paiva B, San Miguel J, Flicek P, Gut I, Melnick A, Mitsiades CS, Licht JD, Campo E, Stunnenberg HG, Agirre X, Prosper F, Martin-Subero JI. Chromatin activation as a unifying principle underlying pathogenic mechanisms in multiple myeloma. Genome Res 2020; 30:1217-1227. [PMID: 32820006 PMCID: PMC7545147 DOI: 10.1101/gr.265520.120] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023]
Abstract
Multiple myeloma (MM) is a plasma cell neoplasm associated with a broad variety of genetic lesions. In spite of this genetic heterogeneity, MMs share a characteristic malignant phenotype whose underlying molecular basis remains poorly characterized. In the present study, we examined plasma cells from MM using a multi-epigenomics approach and demonstrated that, when compared to normal B cells, malignant plasma cells showed an extensive activation of regulatory elements, in part affecting coregulated adjacent genes. Among target genes up-regulated by this process, we found members of the NOTCH, NF-kB, MTOR signaling, and TP53 signaling pathways. Other activated genes included sets involved in osteoblast differentiation and response to oxidative stress, all of which have been shown to be associated with the MM phenotype and clinical behavior. We functionally characterized MM-specific active distant enhancers controlling the expression of thioredoxin (TXN), a major regulator of cellular redox status and, in addition, identified PRDM5 as a novel essential gene for MM. Collectively, our data indicate that aberrant chromatin activation is a unifying feature underlying the malignant plasma cell phenotype.
Collapse
Affiliation(s)
- Raquel Ordoñez
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain
| | - Marta Kulis
- Fundació Clínic per a la Recerca Biomèdica, 08036 Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Nuria Russiñol
- Fundació Clínic per a la Recerca Biomèdica, 08036 Barcelona, Spain
| | - Vicente Chapaprieta
- Departamento de Fundamentos Clínicos, Universitat de Barcelona, 08036 Barcelona, Spain
| | | | - Beatriz García-Torre
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | | | - Guillem Clot
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain.,Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Renée Beekman
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain.,Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Cem Meydan
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medical College, New York, New York 10021, USA
| | - Martí Duran-Ferrer
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Núria Verdaguer-Dot
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Roser Vilarrasa-Blasi
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Paula Soler-Vila
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Leire Garate
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain.,Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Estíbaliz Miranda
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain
| | - Edurne San José-Enériz
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain
| | | | - Teresa Ezponda
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain
| | | | - Amaia Vilas-Zornoza
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain
| | - David Lara-Astiaso
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain
| | - Daphné Dupéré-Richer
- Division of Hematology/Oncology, University of Florida Health Cancer Center, Gainesville, Florida 32610, USA
| | - Joost H A Martens
- Radboud Institute for Molecular Life Sciences, 6525 GA Nijmegen, Netherlands
| | - Halima El-Omri
- Department of Hematology & BMT, Hamad Medical Corporation, NCCCR, Doha, Qatar
| | - Ruba Y Taha
- Department of Hematology & BMT, Hamad Medical Corporation, NCCCR, Doha, Qatar
| | - Maria J Calasanz
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain
| | - Bruno Paiva
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain.,Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Jesus San Miguel
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain.,Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Paul Flicek
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton CB10 1SD, United Kingdom
| | - Ivo Gut
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Ari Melnick
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medical College, New York, New York 10021, USA
| | - Constantine S Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Jonathan D Licht
- Division of Hematology/Oncology, University of Florida Health Cancer Center, Gainesville, Florida 32610, USA
| | - Elias Campo
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain.,Fundació Clínic per a la Recerca Biomèdica, 08036 Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Departamento de Fundamentos Clínicos, Universitat de Barcelona, 08036 Barcelona, Spain
| | | | - Xabier Agirre
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain
| | - Felipe Prosper
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain.,Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Jose I Martin-Subero
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain.,Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Departamento de Fundamentos Clínicos, Universitat de Barcelona, 08036 Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| |
Collapse
|
19
|
Akhtar S, Ali TA, Faiyaz A, Khan OS, Raza SS, Kulinski M, Omri HE, Bhat AA, Uddin S. Cytokine-Mediated Dysregulation of Signaling Pathways in the Pathogenesis of Multiple Myeloma. Int J Mol Sci 2020; 21:5002. [PMID: 32679860 PMCID: PMC7403981 DOI: 10.3390/ijms21145002] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/06/2020] [Accepted: 03/06/2020] [Indexed: 12/15/2022] Open
Abstract
Multiple myeloma (MM) is a hematologic disorder of B lymphocytes characterized by the accumulation of malignant plasma cells (PCs) in the bone marrow. The altered plasma cells overproduce abnormal monoclonal immunoglobulins and also stimulate osteoclasts. The host's immune system and microenvironment are of paramount importance in the growth of PCs and, thus, in the pathogenesis of the disease. The interaction of MM cells with the bone marrow (BM) microenvironment through soluble factors and cell adhesion molecules causes pathogenesis of the disease through activation of multiple signaling pathways, including NF-κβ, PI3K/AKT and JAK/STAT. These activated pathways play a critical role in the inhibition of apoptosis, sustained proliferation, survival and migration of MM cells. Besides, these pathways also participate in developing resistance against the chemotherapeutic drugs in MM. The imbalance between inflammatory and anti-inflammatory cytokines in MM leads to an increased level of pro-inflammatory cytokines, which in turn play a significant role in dysregulation of signaling pathways and proliferation of MM cells; however, the association appears to be inadequate and needs more research. In this review, we are highlighting the recent findings on the roles of various cytokines and growth factors in the pathogenesis of MM and the potential therapeutic utility of aberrantly activated signaling pathways to manage the MM disease.
Collapse
Affiliation(s)
- Sabah Akhtar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (S.A.); (T.A.A.); (A.F.); (M.K.)
| | - Tayyiba A. Ali
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (S.A.); (T.A.A.); (A.F.); (M.K.)
| | - Ammara Faiyaz
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (S.A.); (T.A.A.); (A.F.); (M.K.)
| | - Omar S. Khan
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA;
| | - Syed Shadab Raza
- Department of Stem Cell Biology and Regenerative Medicine, Era University, Lucknow 226003, India;
| | - Michal Kulinski
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (S.A.); (T.A.A.); (A.F.); (M.K.)
| | - Halima El Omri
- National Cancer Care and Research, Hamad Medical Corporation, Doha 3050, Qatar;
| | - Ajaz A. Bhat
- Translational Medicine, Research Branch, Sidra Medicine, Doha 26999, Qatar;
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (S.A.); (T.A.A.); (A.F.); (M.K.)
- Dermatology Institute, Department of Dermatology and Venereology, Hamad Medical Corporation, Doha 3050, Qatar
| |
Collapse
|
20
|
Bai H, Xu P, Wang L, Chen B. A centromere-associated gene score for rapid determination of risk in multiple myeloma. Am J Transl Res 2020; 12:2425-2438. [PMID: 32655781 PMCID: PMC7344103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/25/2020] [Indexed: 06/11/2023]
Abstract
Risk stratification in patients with multiple myeloma (MM) remains a challenge. As clinicopathological characteristics have been proven deficient for accurately defining risk stratification, molecular markers have gradually become the focus of interests. This study investigated the expressions of centromere-associated genes in MM patients, their potential as prognostic markers, and their roles in disease progression. Several cohorts of 2301 MM patients were enrolled and gene expression profiling (GEP) was used to screen for CENP-A through CENP-W. Correlations between centromere-associated genes and clinicopathological characteristics, proliferative activity and recurrence of MM patients were analyzed. Clinically, CENP-E/H/K/L/N/U/W expressions were present at high-risk MM, which were even stronger elevated in patients with high tumor burden and recurrence. Mechanistically, CENP-E/H/K/L/N/U/W and FOXM1 were positively expressed in MM patients, which play synergistic or additive effects in clinical outcome. Furthermore, CENP-E/H/K/L/N/U/W were used to construct a centromere-associated gene score (CGS) model, which proved to be strongly prognostic values in several independent cohorts compared to usual clinical prognostic parameters using multivariate Cox analysis. Patients in the CGS low-risk group were significantly related to better clinical outcome than those in high-risk group. In this study, we provided proof-of-concept that CENP-E/H/K/L/N/U/W have critical roles in MM patients' progression and prognosis. The CGS model validated in different datasets clearly indicated novel risk stratification for personalized anti-MM treatments.
Collapse
Affiliation(s)
- Hua Bai
- Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing 210008, Jiangsu, China
| | - Peipei Xu
- Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing 210008, Jiangsu, China
| | - Lili Wang
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical UniversityNanjing 210008, Jiangsu, China
| | - Bing Chen
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical UniversityNanjing 210008, Jiangsu, China
| |
Collapse
|
21
|
Bolomsky A, Muller J, Stangelberger K, Lejeune M, Duray E, Breid H, Vrancken L, Pfeiffer C, Hübl W, Willheim M, Weetall M, Branstrom A, Zojer N, Caers J, Ludwig H. The anti-mitotic agents PTC-028 and PTC596 display potent activity in pre-clinical models of multiple myeloma but challenge the role of BMI-1 as an essential tumour gene. Br J Haematol 2020; 190:877-890. [PMID: 32232850 DOI: 10.1111/bjh.16595] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 03/02/2020] [Indexed: 12/29/2022]
Abstract
Future progress in the treatment of multiple myeloma (MM) requires both the characterisation of key drivers of the disease and novel, innovative approaches to tackle these vulnerabilities. The present study focussed on the pre-clinical evaluation of a novel drug class, BMI-1 modulators, in MM. We demonstrate potent activity of PTC-028 and PTC596 in a comprehensive set of in vitro and in vivo models, including models of drug resistance and stromal support. Treatment of MM cells with PTC-028 and PTC596 downregulated BMI-1 protein levels, which was found to correlate with drug activity. Surprisingly, BMI-1 was dispensable for the activity of BMI-1 modulators and MM cell growth. Our data rather point to mitotic arrest accompanied by myeloid cell leukaemia-1 (MCL-1) loss as key anti-MM mechanisms and reveal impaired MYC and AKT signalling activity due to BMI-1 modulator treatment. Moreover, we observed a complete eradication of MM after PTC596 treatment in the 5TGM.1 in vivo model and define epigenetic compounds and B cell leukaemia/lymphoma 2 homology domain 3 (BH3) mimetics as promising combination partners. These results bring into question the postulated role of BMI-1 as an essential MM gene and confirm BMI-1 modulators as potent anti-mitotic agents with encouraging pre-clinical activity that supports their rapid translation into clinical trials.
Collapse
Affiliation(s)
- Arnold Bolomsky
- Department of Medicine I, Wilhelminen Cancer Research Institute, Wilhelminenspital, Vienna, Austria
| | - Joséphine Muller
- Laboratory of Hematology, GIGA-I3, University of Liège, Liège, Belgium
| | - Kathrin Stangelberger
- Department of Medicine I, Wilhelminen Cancer Research Institute, Wilhelminenspital, Vienna, Austria
| | - Margaux Lejeune
- Laboratory of Hematology, GIGA-I3, University of Liège, Liège, Belgium
| | - Elodie Duray
- Laboratory of Hematology, GIGA-I3, University of Liège, Liège, Belgium
| | - Helene Breid
- Department of Medicine I, Wilhelminen Cancer Research Institute, Wilhelminenspital, Vienna, Austria
| | - Louise Vrancken
- Laboratory of Hematology, GIGA-I3, University of Liège, Liège, Belgium
| | - Christina Pfeiffer
- Department of Medicine I, Wilhelminen Cancer Research Institute, Wilhelminenspital, Vienna, Austria
| | - Wolfgang Hübl
- Department of Laboratory Medicine, Wilhelminenspital, Vienna, Austria
| | - Martin Willheim
- Department of Laboratory Medicine, Wilhelminenspital, Vienna, Austria
| | | | | | - Niklas Zojer
- Department of Medicine I, Wilhelminen Cancer Research Institute, Wilhelminenspital, Vienna, Austria
| | - Jo Caers
- Laboratory of Hematology, GIGA-I3, University of Liège, Liège, Belgium
| | - Heinz Ludwig
- Department of Medicine I, Wilhelminen Cancer Research Institute, Wilhelminenspital, Vienna, Austria
| |
Collapse
|
22
|
Chatonnet F, Pignarre A, Sérandour AA, Caron G, Avner S, Robert N, Kassambara A, Laurent A, Bizot M, Agirre X, Prosper F, Martin-Subero JI, Moreaux J, Fest T, Salbert G. The hydroxymethylome of multiple myeloma identifies FAM72D as a 1q21 marker linked to proliferation. Haematologica 2020; 105:774-783. [PMID: 31221779 PMCID: PMC7049362 DOI: 10.3324/haematol.2019.222133] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/19/2019] [Indexed: 01/16/2023] Open
Abstract
Cell identity relies on the cross-talk between genetics and epigenetics and their impact on gene expression. Oxidation of 5-methylcytosine (5mC) into 5-hydroxymethylcytosine (5hmC) is the first step of an active DNA demethylation process occurring mainly at enhancers and gene bodies and, as such, participates in processes governing cell identity in normal and pathological conditions. Although genetic alterations are well documented in multiple myeloma (MM), epigenetic alterations associated with this disease have not yet been thoroughly analyzed. To gain insight into the biology of MM, genome-wide 5hmC profiles were obtained and showed that regions enriched in this modified base overlap with MM enhancers and super enhancers and are close to highly expressed genes. Through the definition of a MM-specific 5hmC signature, we identified FAM72D as a poor prognostic gene located on 1q21, a region amplified in high risk myeloma. We further uncovered that FAM72D functions as part of the FOXM1 transcription factor network controlling cell proliferation and survival and we evidenced an increased sensitivity of cells expressing high levels of FOXM1 and FAM72 to epigenetic drugs targeting histone deacetylases and DNA methyltransferases.
Collapse
Affiliation(s)
- Fabrice Chatonnet
- Université Rennes 1, Établissement Français du Sang de Bretaggne, Inserm, MICMAC -UMR_S 1236, Rennes, France
- Laboratoire d'Hématologie, Pôle de Biologie, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Amandine Pignarre
- Université Rennes 1, Établissement Français du Sang de Bretaggne, Inserm, MICMAC -UMR_S 1236, Rennes, France
- Laboratoire d'Hématologie, Pôle de Biologie, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Aurélien A Sérandour
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
- Ecole Centrale de Nantes, Nantes, France
- Institut de Cancérologie de l'Ouest, Site René-Gauducheau, Saint-Herblain, France
| | - Gersende Caron
- Université Rennes 1, Établissement Français du Sang de Bretaggne, Inserm, MICMAC -UMR_S 1236, Rennes, France
- Laboratoire d'Hématologie, Pôle de Biologie, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Stéphane Avner
- SPARTE, IGDR, CNRS UMR6290, University Rennes 1, Rennes, France
| | - Nicolas Robert
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
| | | | - Audrey Laurent
- SPARTE, IGDR, CNRS UMR6290, University Rennes 1, Rennes, France
| | - Maud Bizot
- SPARTE, IGDR, CNRS UMR6290, University Rennes 1, Rennes, France
| | - Xabier Agirre
- Area de Oncología, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Felipe Prosper
- Area de Oncología, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | | | - Jérôme Moreaux
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
- IGH, CNRS, Univ Montpellier, France
| | - Thierry Fest
- Université Rennes 1, Établissement Français du Sang de Bretaggne, Inserm, MICMAC -UMR_S 1236, Rennes, France
- Laboratoire d'Hématologie, Pôle de Biologie, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Gilles Salbert
- SPARTE, IGDR, CNRS UMR6290, University Rennes 1, Rennes, France
| |
Collapse
|
23
|
Sheng Y, Yu C, Liu Y, Hu C, Ma R, Lu X, Ji P, Chen J, Mizukawa B, Huang Y, Licht JD, Qian Z. FOXM1 regulates leukemia stem cell quiescence and survival in MLL-rearranged AML. Nat Commun 2020; 11:928. [PMID: 32066721 PMCID: PMC7026046 DOI: 10.1038/s41467-020-14590-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 12/16/2019] [Indexed: 12/16/2022] Open
Abstract
FOXM1, a known transcription factor, promotes cell proliferation in a variety of cancer cells. Here we show that Foxm1 is required for survival, quiescence and self-renewal of MLL-AF9 (MA9)-transformed leukemia stem cells (LSCs) in vivo. Mechanistically, Foxm1 upregulation activates the Wnt/β-catenin signaling pathways by directly binding to β-catenin and stabilizing β-catenin protein through inhibiting its degradation, thereby preserving LSC quiescence, and promoting LSC self-renewal in MLL-rearranged AML. More importantly, inhibition of FOXM1 markedly suppresses leukemogenic potential and induces apoptosis of primary LSCs from MLL-rearranged AML patients in vitro and in vivo in xenograft mice. Thus, our study shows a critical role and mechanisms of Foxm1 in MA9-LSCs, and indicates that FOXM1 is a potential therapeutic target for selectively eliminating LSCs in MLL-rearranged AML.
Collapse
Affiliation(s)
- Yue Sheng
- Division of Hematology/Oncology, UF Health Cancer Center, University of Florida, Gainesville, FL, USA
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Chunjie Yu
- Division of Hematology/Oncology, UF Health Cancer Center, University of Florida, Gainesville, FL, USA
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Yin Liu
- Division of Hematology/Oncology, UF Health Cancer Center, University of Florida, Gainesville, FL, USA
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Chao Hu
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Rui Ma
- Institute for Tuberculosis Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Xinyan Lu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, IL, USA
| | - Peng Ji
- Department of Pathology, Feinberg School of Medicine, Northwestern University, IL, USA
| | - Jianjun Chen
- Department of System Biology, City of Hope, CA, USA
| | - Benjamin Mizukawa
- Cancer & Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Yong Huang
- Department of Medicine, University of Virginia, Charlottestville, VA, USA
| | - Jonathan D Licht
- Division of Hematology/Oncology, UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Zhijian Qian
- Division of Hematology/Oncology, UF Health Cancer Center, University of Florida, Gainesville, FL, USA.
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
24
|
Bai H, Chen B. BAG3 regulates multiple myeloma cell proliferation through FOXM1/Rb/E2F axis. Cancer Gene Ther 2019; 27:108-111. [PMID: 31801989 DOI: 10.1038/s41417-019-0154-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/26/2019] [Indexed: 01/03/2023]
Affiliation(s)
- Hua Bai
- Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| | - Bing Chen
- Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| |
Collapse
|
25
|
Stefka AT, Johnson D, Rosebeck S, Park JH, Nakamura Y, Jakubowiak AJ. Potent anti-myeloma activity of the TOPK inhibitor OTS514 in pre-clinical models. Cancer Med 2019; 9:324-334. [PMID: 31714026 PMCID: PMC6943155 DOI: 10.1002/cam4.2695] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/30/2019] [Accepted: 10/25/2019] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) continues to be considered incurable, necessitating new drug discovery. The mitotic kinase T‐LAK cell‐originated protein kinase/PDZ‐binding kinase (TOPK/PBK) is associated with proliferation of tumor cells, maintenance of cancer stem cells, and poor patient prognosis in many cancers. In this report, we demonstrate potent anti‐myeloma effects of the TOPK inhibitor OTS514 for the first time. OTS514 induces cell cycle arrest and apoptosis at nanomolar concentrations in a series of human myeloma cell lines (HMCL) and prevents outgrowth of a putative CD138+ stem cell population from MM patient‐derived peripheral blood mononuclear cells. In bone marrow cells from MM patients, OTS514 treatment exhibited preferential killing of the malignant CD138+ plasma cells compared with the CD138− compartment. In an aggressive mouse xenograft model, OTS964 given orally at 100 mg/kg 5 days per week was well tolerated and reduced tumor size by 48%‐81% compared to control depending on the initial graft size. FOXO3 and its transcriptional targets CDKN1A (p21) and CDKN1B (p27) were elevated and apoptosis was induced with OTS514 treatment of HMCLs. TOPK inhibition also induced loss of FOXM1 and disrupted AKT, p38 MAPK, and NF‐κB signaling. The effects of OTS514 were independent of p53 mutation or deletion status. Combination treatment of HMCLs with OTS514 and lenalidomide produced synergistic effects, providing a rationale for the evaluation of TOPK inhibition in existing myeloma treatment regimens.
Collapse
Affiliation(s)
- Andrew T Stefka
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - David Johnson
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Shaun Rosebeck
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Jae-Hyun Park
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Yusuke Nakamura
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | | |
Collapse
|
26
|
Vlummens P, De Veirman K, Menu E, De Bruyne E, Offner F, Vanderkerken K, Maes K. The Use of Murine Models for Studying Mechanistic Insights of Genomic Instability in Multiple Myeloma. Front Genet 2019; 10:740. [PMID: 31475039 PMCID: PMC6704229 DOI: 10.3389/fgene.2019.00740] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) is a B-cell malignancy characterized by the accumulation of clonal plasma cells in the bone marrow. In normal plasma cell development, cells undergo programmed DNA breaks and translocations, a process necessary for generation of a wide repertoire of antigen-specific antibodies. This process also makes them vulnerable for the acquisition of chromosomal defects. Well-known examples of these aberrations, already seen at time of MM diagnosis, are hyperdiploidy or the translocations involving the immunoglobulin heavy chain. Over the recent years, however, novel aspects concerning genomic instability and its role in tumor development, disease progression and nascence of refractory disease were identified. As such, genomic instability is becoming a very relevant research topic with the potential identification of novel disease pathways. In this review, we aim to describe recent studies involving murine MM models focusing on the deregulation of processes implicated in genomic instability and their clinical impact. More specifically, we will discuss chromosomal instability, DNA damage and repair responses, development of drug resistance, and recent insights into the study of clonal hierarchy using different murine MM models. Lastly, we will discuss the importance and the use of murine MM models in the pre-clinical evaluation of promising novel therapeutic agents.
Collapse
Affiliation(s)
- Philip Vlummens
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium.,Department of Clinical Hematology, Ghent University Hospital, Gent, Belgium
| | - Kim De Veirman
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eline Menu
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Elke De Bruyne
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Fritz Offner
- Department of Clinical Hematology, Ghent University Hospital, Gent, Belgium
| | - Karin Vanderkerken
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ken Maes
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
27
|
Haque M, Li J, Huang YH, Almowaled M, Barger CJ, Karpf AR, Wang P, Chen W, Turner SD, Lai R. NPM-ALK Is a Key Regulator of the Oncoprotein FOXM1 in ALK-Positive Anaplastic Large Cell Lymphoma. Cancers (Basel) 2019; 11:E1119. [PMID: 31390744 PMCID: PMC6721812 DOI: 10.3390/cancers11081119] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/23/2019] [Accepted: 07/29/2019] [Indexed: 12/11/2022] Open
Abstract
Forkhead Box M1 (FOXM1) is an oncogenic transcription factor implicated in the pathogenesis of solid and hematologic cancers. In this study, we examined the significance of FOXM1 in NPM-ALK-positive anaplastic large cell lymphoma (NPM-ALK + ALCL), with a focus on how it interacts with NPM-ALK, which is a key oncogenic driver in these tumors. FOXM1 was expressed in NPM-ALK + ALCL cell lines (5/5), patient samples (21/21), and tumors arising in NPM-ALK transgenic mice (4/4). FOXM1 was localized in the nuclei and confirmed to be transcriptionally active. Inhibition of FOXM1 in two NPM-ALK + ALCL cells using shRNA and pharmalogic agent (thiostrepton) resulted in reductions in cell growth and soft-agar colony formation, which were associated with apoptosis and cell-cycle arrest. FOXM1 is functionally linked to NPM-ALK, as FOXM1 enhanced phosphorylation of the NPM-ALK/STAT3 axis. Conversely, DNA binding and transcriptional activity of FOXM1 was dependent on the expression of NPM-ALK. Further studies showed that this dependency hinges on the binding of FOXM1 to NPM1 that heterodimerizes with NPM-ALK, and the phosphorylation status of NPM-ALK. In conclusion, we identified FOXM1 as an important oncogenic protein in NPM-ALK+ ALCL. Our results exemplified that NPM-ALK exerts oncogenic effects in the nuclei and illustrated a novel role of NPM1 in NPM-ALK pathobiology.
Collapse
Affiliation(s)
- Moinul Haque
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Jing Li
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G2R3, Canada
- Electron Microscopy Center, Basic Medical Science College, Harbin Medical University, Harbin 150080, Heilongjiang, China
| | - Yung-Hsing Huang
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Meaad Almowaled
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Carter J Barger
- Eppley Institute and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Adam R Karpf
- Eppley Institute and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Peng Wang
- Department of Hematology, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Will Chen
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Suzanne D Turner
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge CB20QQ, UK
| | - Raymond Lai
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G2R3, Canada.
- Department of Oncology, University of Alberta, Edmonton, AB T6G2R3, Canada.
| |
Collapse
|
28
|
Akbari V, Kallhor M, Mollashahi B, Movafagh A. Transcriptome and Network Dissection of Microsatellite Stable and Highly Instable Colorectal Cancer. Asian Pac J Cancer Prev 2019; 20:2445-2454. [PMID: 31450919 PMCID: PMC6852802 DOI: 10.31557/apjcp.2019.20.8.2445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Indexed: 12/24/2022] Open
Abstract
Background: Colorectal cancer (CRC) is one of the most common cancers worldwide with high number of mortality every year. Microsatellite instability (MSI) is a considerable feature of CRC which affects prognosis and treatment. High level of MSI or MSI-high (MSI-H) colorectal cancer has better prognosis and immunotherapy response, while microsatellite stable (MSS) CRC has better response to 5-fluorouracil (5-FU)-based chemotherapy. More studies are needed, specifically on MSS CRC which has worse prognosis, to further reveal biological differences and similarities between MSS and MSI colorectal cancer, which may equip us with the knowledge to develop more promising therapeutic approaches to target both types or be more effective for each type. Methods: We aimed to find affected biological processes and their regulators in both type, MSS and MSI-H, of CRC; as well as reveal specific ones in each type. We applied meta- and network analysis on freely available transcriptome data in MSS and MSI-H colorectal cancer from gene expression omnibus (GEO) database to detect common differentially expressed genes (DEGs) and critical biological processes and predict their most significant regulators. Results: Our results demonstrate considerable up and downregulation in cell cycle and lipid catabolism processes, respectively; and introduced MYC and FOXM1 as two central and up-stream regulators of DEGs in both type of CRC. Chemokine-mediated processes displayed up-regulation in MSI-H type, while metastasis-related processes showed more activation in MSS CRC. Additionally, DACH1 and TP53 were detected as two important transcription factors that differentially expressed just in MSS and MSI-H, respectively. Conclusion: Our results can explain why MSI and MSS CRC display different immunotherapy response, prognosis, and metastasis feature. Moreover, our predicted up-stream regulators in the regulatory networks may be promising therapeutic targets.
Collapse
Affiliation(s)
- Vahid Akbari
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC Canada. ,
| | - Marzieh Kallhor
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Behrouz Mollashahi
- Department of Cellular-Molecular Biology, Faculty of Biological Sciences and Technologies Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolfazl Movafagh
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Wang W, Wei R, Liu S, Qiao L, Hou J, Gu C, Yang Y. BTK induces CAM-DR through regulation of CXCR4 degradation in multiple myeloma. Am J Transl Res 2019; 11:4139-4150. [PMID: 31396324 PMCID: PMC6684885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 06/09/2019] [Indexed: 06/10/2023]
Abstract
Cellular adhesion-mediated drug resistance (CAM-DR) occurs frequently in patients with relapsed or refractory multiple myeloma (MM). Elucidating the mechanism underlying CAM-DR and developing the corresponding treatment may prove to be promising for the clinical management of MM. Bruton's tyrosine kinase (BTK) has been attracting attention in relation to MM progression and drug resistance. BTK was reported to be associated with cell surface CXCR4, a classic cell adhesion molecule and homing factor. However, the exact association between BTK and CAM-DR in MM remains elusive. In this study, we demonstrated that promoting BTK expression induced MM cell adherence to the extracellular matrix (ECM) and stromal cells in vitro and in vivo, and that CAM-DR could be reversed by separating MM cells from ECM or stromal cells. Enhancing BTK expression levels increased CXCR4 expression in MM cells. In addition, BTK may bind directly with CXCR4 and prevent its ubiquitination-induced degradation. Finally, a BTK inhibitor exerted synergistic therapeutic effects with bortezomib in a 5TMM3VT MM mouse model. These findings revealed a novel role of BTK in CAM-DR and may provide a promising approach to MM treatment.
Collapse
Affiliation(s)
- Wang Wang
- School of Medicine and Life Sciences, Nanjing University of Chinese MedicineNanjing 210023, Jiangsu, China
- The Third Affiliated Hospital of Nanjing University of Chinese MedicineNanjing 210001, Jiangsu, China
| | - Rongfang Wei
- School of Medicine and Life Sciences, Nanjing University of Chinese MedicineNanjing 210023, Jiangsu, China
| | - Shijia Liu
- Affiliated Hospital of Nanjing University of Chinese MedicineNanjing 210029, Jiangsu, China
| | - Li Qiao
- School of Medicine and Life Sciences, Nanjing University of Chinese MedicineNanjing 210023, Jiangsu, China
| | - Jianhao Hou
- School of Medicine and Life Sciences, Nanjing University of Chinese MedicineNanjing 210023, Jiangsu, China
| | - Chunyan Gu
- School of Medicine and Life Sciences, Nanjing University of Chinese MedicineNanjing 210023, Jiangsu, China
- The Third Affiliated Hospital of Nanjing University of Chinese MedicineNanjing 210001, Jiangsu, China
| | - Ye Yang
- School of Medicine and Life Sciences, Nanjing University of Chinese MedicineNanjing 210023, Jiangsu, China
- School of Holistic Integrative Medicine, Nanjing University of Chinese MedicineNanjing 210023, Jiangsu, China
| |
Collapse
|
30
|
Haroon UH, Davis NF, Mohan P, Little DM, Smyth G, Forde JC, Power RE. Incidence, Management, and Clinical Outcomes of Prostate Cancer in Kidney Transplant Recipients. EXP CLIN TRANSPLANT 2019; 17:298-303. [DOI: 10.6002/ect.2018.0048] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
31
|
Gu C, Jing X, Holman C, Sompallae R, Zhan F, Tricot G, Yang Y, Janz S. Upregulation of FOXM1 leads to diminished drug sensitivity in myeloma. BMC Cancer 2018; 18:1152. [PMID: 30463534 PMCID: PMC6249818 DOI: 10.1186/s12885-018-5015-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 10/30/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Following up on previous work demonstrating the involvement of the transcription factor forkhead box M1 (FOXM1) in the biology and outcome of a high-risk subset of newly diagnosed multiple myeloma (nMM), this study evaluated whether FOXM1 gene expression may be further upregulated upon tumor recurrence in patients with relapsed multiple myeloma (rMM). Also assessed was the hypothesis that increased levels of FOXM1 diminish the sensitivity of myeloma cells to commonly used myeloma drugs, such as the proteasome inhibitor bortezomib (Bz) and the DNA intercalator doxorubicin (Dox). METHODS FOXM1 message was evaluated in 88 paired myeloma samples from patients with nMM and rMM, using gene expression microarrays as measurement tool. Sources of differential gene expression were identified and outlier analyses were performed using statistical methods. Two independent human myeloma cell lines (HMCLs) containing normal levels of FOXM1 (FOXM1N) or elevated levels of lentivirus-encoded FOXM1 (FOXM1Hi) were employed to determine FOXM1-dependent changes in cell proliferation, survival, efflux-pump activity, and drug sensitivity. Levels of retinoblastoma (Rb) protein were determined with the assistance of Western blotting. RESULTS Upregulation of FOXM1 occurred in 61 of 88 (69%) patients with rMM, including 4 patients that exhibited > 20-fold elevated expression peaks. Increased FOXM1 levels in FOXM1Hi myeloma cells caused partial resistance to Bz (1.9-5.6 fold) and Dox (1.5-2.9 fold) in vitro, using FOXM1N myeloma as control. Reduced sensitivity of FOXM1Hi cells to Bz was confirmed in vivo using myeloma-in-mouse xenografts. FOXM1-dependent regulation of total and phosphorylated Rb agreed with a working model of myeloma suggesting that FOXM1 governs both chromosomal instability (CIN) and E2F-dependent proliferation, using a mechanism that involves interaction with NIMA related kinase 2 (NEK2) and cyclin dependent kinase 6 (CDK6), respectively. CONCLUSIONS These findings enhanced our understanding of the emerging FOXM1 genetic network in myeloma and provided preclinical support for the therapeutic targeting of the FOXM1-NEK2 and CDK4/6-Rb-E2F pathways using small-drug CDK and NEK2 inhibitors. Clinical research is warranted to assess whether this approach may overcome drug resistance in FOXM1Hi myeloma and, thereby, improve the outcome of patients in which the transcription factor is expressed at high levels.
Collapse
Affiliation(s)
- Chunyan Gu
- The Third Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, 210023 China
- Department of Pathology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa 52242 USA
| | - Xuefang Jing
- Department of Pathology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa 52242 USA
| | - Carol Holman
- Department of Pathology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa 52242 USA
| | - Ramakrishna Sompallae
- Department of Pathology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa 52242 USA
- Iowa Institute for Genetics, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa 52242 USA
| | - Fenghuang Zhan
- Department of Internal Medicine, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa 52242 USA
- Holden Comprehensive Cancer Center, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa 52242 USA
| | - Guido Tricot
- Department of Internal Medicine, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa 52242 USA
- Holden Comprehensive Cancer Center, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa 52242 USA
| | - Ye Yang
- The Third Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, 210023 China
- Key Laboratory of Acupuncture and Medicine Research, Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023 China
| | - Siegfried Janz
- Department of Pathology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa 52242 USA
- Holden Comprehensive Cancer Center, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa 52242 USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53213 USA
| |
Collapse
|
32
|
Muller J, Bolomsky A, Dubois S, Duray E, Stangelberger K, Plougonven E, Lejeune M, Léonard A, Marty C, Hempel U, Baron F, Beguin Y, Cohen-Solal M, Ludwig H, Heusschen R, Caers J. Maternal embryonic leucine zipper kinase inhibitor OTSSP167 has preclinical activity in multiple myeloma bone disease. Haematologica 2018; 103:1359-1368. [PMID: 29748441 PMCID: PMC6068043 DOI: 10.3324/haematol.2017.185397] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 05/03/2018] [Indexed: 11/09/2022] Open
Abstract
Multiple myeloma bone disease is characterized by an uncoupling of bone remodeling in the multiple myeloma microenvironment, resulting in the development of lytic bone lesions. Most myeloma patients suffer from these bone lesions, which not only cause morbidity but also negatively impact survival. The development of novel therapies, ideally with a combined anti-resorptive and bone-anabolic effect, is of great interest because lesions persist with the current standard of care, even in patients in complete remission. We have previously shown that MELK plays a central role in proliferation-associated high-risk multiple myeloma and its inhibition with OTSSP167 resulted in decreased tumor load. MELK inhibition in bone cells has not yet been explored, although some reports suggest that factors downstream of MELK stimulate osteoclast activity and inhibit osteoblast activity, which makes MELK inhibition a promising therapeutic approach. Therefore, we assessed the effect of OTSSP167 on bone cell activity and the development of myeloma-induced bone disease. OTSSP167 inhibited osteoclast activity in vitro by decreasing progenitor viability as well as via a direct anti-resorptive effect on mature osteoclasts. In addition, OTSSP167 stimulated matrix deposition and mineralization by osteoblasts in vitro. This combined anti-resorptive and osteoblast-stimulating effect of OTSSP167 resulted in the complete prevention of lytic lesions and bone loss in myeloma-bearing mice. Immunohistomorphometric analyses corroborated our in vitro findings. In conclusion, we show that OTSSP167 has a direct effect on myeloma-induced bone disease in addition to its anti-multiple myeloma effect, which warrants further clinical development of MELK inhibition in multiple myeloma.
Collapse
Affiliation(s)
| | - Arnold Bolomsky
- Wilhelminen Cancer Research Institute, Department of Medicine I, Wilhelminenspital, Vienna, Austria
| | - Sophie Dubois
- Laboratory of Hematology, GIGA-I3, University of Liège, Belgium
| | - Elodie Duray
- Laboratory of Hematology, GIGA-I3, University of Liège, Belgium
| | - Kathrin Stangelberger
- Wilhelminen Cancer Research Institute, Department of Medicine I, Wilhelminenspital, Vienna, Austria
| | - Erwan Plougonven
- PEPs (Products, Environments, Processes), Chemical Engineering, Liège, Belgium
| | - Margaux Lejeune
- Laboratory of Hematology, GIGA-I3, University of Liège, Belgium
| | - Angélique Léonard
- PEPs (Products, Environments, Processes), Chemical Engineering, Liège, Belgium
| | | | - Ute Hempel
- Institute of Physiological Chemistry, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Frédéric Baron
- Laboratory of Hematology, GIGA-I3, University of Liège, Belgium.,Department of Hematology, CHU de Liège, Belgium
| | - Yves Beguin
- Laboratory of Hematology, GIGA-I3, University of Liège, Belgium.,Department of Hematology, CHU de Liège, Belgium
| | | | - Heinz Ludwig
- Wilhelminen Cancer Research Institute, Department of Medicine I, Wilhelminenspital, Vienna, Austria
| | - Roy Heusschen
- Laboratory of Hematology, GIGA-I3, University of Liège, Belgium
| | - Jo Caers
- Laboratory of Hematology, GIGA-I3, University of Liège, Belgium .,Department of Hematology, CHU de Liège, Belgium
| |
Collapse
|
33
|
Upregulation of FOXM1 in a subset of relapsed myeloma results in poor outcome. Blood Cancer J 2018; 8:22. [PMID: 29449574 PMCID: PMC5814454 DOI: 10.1038/s41408-018-0060-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 11/18/2017] [Accepted: 12/11/2017] [Indexed: 12/18/2022] Open
|
34
|
Bolomsky A, Heusschen R, Schlangen K, Stangelberger K, Muller J, Schreiner W, Zojer N, Caers J, Ludwig H. Maternal embryonic leucine zipper kinase is a novel target for proliferation-associated high-risk myeloma. Haematologica 2017; 103:325-335. [PMID: 29122991 PMCID: PMC5792277 DOI: 10.3324/haematol.2017.172973] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 10/27/2017] [Indexed: 01/14/2023] Open
Abstract
Treatment of high-risk patients is a major challenge in multiple myeloma. This is especially true for patients assigned to the gene expression profiling-defined proliferation subgroup. Although recent efforts have identified some key players of proliferative myeloma, genetic interactions and players that can be targeted with clinically effective drugs have to be identified in order to overcome the poor prognosis of these patients. We therefore examined maternal embryonic leucine zipper kinase (MELK) for its implications in hyper-proliferative myeloma and analyzed the activity of the MELK inhibitor OTSSP167 both in vitro and in vivoMELK was found to be significantly overexpressed in the proliferative subgroup of myeloma. This finding translated into poor overall survival in patients with high vs low MELK expression. Enrichment analysis of upregulated genes in myeloma cells of MELKhigh patients confirmed the strong implications in myeloma cell proliferation. Targeting MELK with OTSSP167 impaired the growth and survival of myeloma cells, thereby affecting central survival factors such as MCL-1 and IRF4 This activity was also observed in the 5TGM.1 murine model of myeloma. OTSSP167 reduced bone marrow infiltration and serum paraprotein levels in a dose-dependent manner. In addition, we revealed a strong link between MELK and other proliferation-associated high-risk genes (PLK-1, EZH2, FOXM1, DEPDC1) and MELK inhibition also impaired the expression of those genes. We therefore conclude that MELK is an essential component of a proliferative gene signature and that pharmacological inhibition of MELK represents an attractive novel approach to overcome the poor prognosis of high-risk patients with a proliferative expression pattern.
Collapse
Affiliation(s)
- Arnold Bolomsky
- Wilhelminen Cancer Research Institute, Department of Medicine I, Wilhelminenspital, Vienna, Austria
| | - Roy Heusschen
- Laboratory of Hematology, GIGA-I3, University of Liège, Belgium
| | - Karin Schlangen
- Center for Medical Statistics, Informatics and Intelligent Systems, Section for Biosimulation and Bioinformatics, Medical University of Vienna, Austria and
| | - Kathrin Stangelberger
- Wilhelminen Cancer Research Institute, Department of Medicine I, Wilhelminenspital, Vienna, Austria
| | | | - Wolfgang Schreiner
- Center for Medical Statistics, Informatics and Intelligent Systems, Section for Biosimulation and Bioinformatics, Medical University of Vienna, Austria and
| | - Niklas Zojer
- Wilhelminen Cancer Research Institute, Department of Medicine I, Wilhelminenspital, Vienna, Austria
| | - Jo Caers
- Laboratory of Hematology, GIGA-I3, University of Liège, Belgium .,Division of Hematology, Department of Medicine, University and CHU of Liège, Belgium
| | - Heinz Ludwig
- Wilhelminen Cancer Research Institute, Department of Medicine I, Wilhelminenspital, Vienna, Austria
| |
Collapse
|
35
|
Egawa M, Yoshida Y, Ogura S, Kurahashi T, Kizu T, Furuta K, Kamada Y, Chatani N, Hamano M, Kiso S, Hikita H, Tatsumi T, Eguchi H, Nagano H, Doki Y, Mori M, Takehara T. Increased expression of Forkhead box M1 transcription factor is associated with clinicopathological features and confers a poor prognosis in human hepatocellular carcinoma. Hepatol Res 2017; 47:1196-1205. [PMID: 28002884 DOI: 10.1111/hepr.12854] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 09/29/2016] [Accepted: 12/19/2016] [Indexed: 02/08/2023]
Abstract
AIM Forkhead Box M1 (FoxM1) is a proliferation-specific transcription factor. In this study, we aimed to elucidate the clinicopathological and prognostic values of FoxM1 expression in human hepatocellular carcinoma (HCC) and correlate FoxM1 expression with various etiologies of liver diseases. We also investigated its therapeutic value in HCC. METHODS We investigated the expression of FoxM1 in tumor tissues and adjacent non-tumor tissues of 79 Japanese HCC patients by quantitative real-time reverse transcription-polymerase chain reaction analysis. Depletion by siRNA or specific inhibition by siomycin A were also used to investigate the effect of FoxM1 inhibition on stem-like features of human HCC cells. RESULTS Quantitative real-time reverse transcription-polymerase chain reaction analysis showed that tumor tissues displayed an approximately 14-fold increase in FoxM1 expression compared with adjacent non-tumor tissues. Interestingly, the expression levels of FoxM1in tumor tissues did not depend on the etiology of liver disease. The expression of FoxM1 in tumor tissues was associated with serum α-fetoprotein level, maximum tumor size, histological grade, TNM staging, and portal involvement. Kaplan-Meier analysis indicated that the high FoxM1 expression (≥median) group had a poor prognosis compared with the low FoxM1 expression (<median) group. Using multivariate analysis, the expression of FoxM1 in tumor tissues was shown to be an independent prognostic factor that affected overall survival and disease-free survival. Furthermore, FoxM1 inhibition by siRNA or siomycin A reduced spheroid colony formation of HCC cells in vitro. CONCLUSION Our data suggest that FoxM1 might be a prognostic biomarker and a promising therapeutic target for HCC.
Collapse
Affiliation(s)
- Mayumi Egawa
- Department of Gastroenterology and Hepatology, Osaka, Japan
| | - Yuichi Yoshida
- Department of Gastroenterology and Hepatology, Osaka, Japan
| | - Satoshi Ogura
- Department of Gastroenterology and Hepatology, Osaka, Japan
| | | | - Takashi Kizu
- Department of Gastroenterology and Hepatology, Osaka, Japan
| | | | - Yoshihiro Kamada
- Department of Gastroenterology and Hepatology, Osaka, Japan.,Department of Molecular Biochemistry and Clinical Investigation, Osaka, Japan
| | | | - Mina Hamano
- Department of Gastroenterology and Hepatology, Osaka, Japan
| | - Shinichi Kiso
- Department of Gastroenterology and Hepatology, Osaka, Japan
| | - Hayato Hikita
- Department of Gastroenterology and Hepatology, Osaka, Japan
| | | | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroaki Nagano
- Department of Digestive Surgery and Surgical Oncology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | | |
Collapse
|
36
|
Nandi D, Cheema PS, Jaiswal N, Nag A. FoxM1: Repurposing an oncogene as a biomarker. Semin Cancer Biol 2017; 52:74-84. [PMID: 28855104 DOI: 10.1016/j.semcancer.2017.08.009] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 08/08/2017] [Accepted: 08/23/2017] [Indexed: 12/16/2022]
Abstract
The past few decades have witnessed a tremendous progress in understanding the biology of cancer, which has led to more comprehensive approaches for global gene expression profiling and genome-wide analysis. This has helped to determine more sophisticated prognostic and predictive signature markers for the prompt diagnosis and precise screening of cancer patients. In the search for novel biomarkers, there has been increased interest in FoxM1, an extensively studied transcription factor that encompasses most of the hallmarks of malignancy. Considering the attractive potential of this multifarious oncogene, FoxM1 has emerged as an important molecule implicated in initiation, development and progression of cancer. Bolstered with the skill to maneuver the proliferation signals, FoxM1 bestows resistance to contemporary anti-cancer therapy as well. This review sheds light on the large body of literature that has accumulated in recent years that implies that FoxM1 neoplastic functions can be used as a novel predictive, prognostic and therapeutic marker for different cancers. This assessment also highlights the key features of FoxM1 that can be effectively harnessed to establish FoxM1 as a strong biomarker in diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Deeptashree Nandi
- Department of Biochemistry, University of Delhi South Campus, New Delhi, 110021, India
| | - Pradeep Singh Cheema
- Department of Biochemistry, University of Delhi South Campus, New Delhi, 110021, India
| | - Neha Jaiswal
- Department of Biochemistry, University of Delhi South Campus, New Delhi, 110021, India
| | - Alo Nag
- Department of Biochemistry, University of Delhi South Campus, New Delhi, 110021, India.
| |
Collapse
|
37
|
Logic programming reveals alteration of key transcription factors in multiple myeloma. Sci Rep 2017; 7:9257. [PMID: 28835615 PMCID: PMC5569101 DOI: 10.1038/s41598-017-09378-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 07/25/2017] [Indexed: 01/24/2023] Open
Abstract
Innovative approaches combining regulatory networks (RN) and genomic data are needed to extract biological information for a better understanding of diseases, such as cancer, by improving the identification of entities and thereby leading to potential new therapeutic avenues. In this study, we confronted an automatically generated RN with gene expression profiles (GEP) from a cohort of multiple myeloma (MM) patients and normal individuals using global reasoning on the RN causality to identify key-nodes. We modeled each patient by his or her GEP, the RN and the possible automatically detected repairs needed to establish a coherent flow of the information that explains the logic of the GEP. These repairs could represent cancer mutations leading to GEP variability. With this reasoning, unmeasured protein states can be inferred, and we can simulate the impact of a protein perturbation on the RN behavior to identify therapeutic targets. We showed that JUN/FOS and FOXM1 activities are altered in almost all MM patients and identified two survival markers for MM patients. Our results suggest that JUN/FOS-activation has a strong impact on the RN in view of the whole GEP, whereas FOXM1-activation could be an interesting way to perturb an MM subgroup identified by our method.
Collapse
|
38
|
Wang J, Li L, Liu S, Zhao Y, Wang L, Du G. FOXC1 promotes melanoma by activating MST1R/PI3K/AKT. Oncotarget 2016; 7:84375-84387. [PMID: 27533251 PMCID: PMC5356666 DOI: 10.18632/oncotarget.11224] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/19/2016] [Indexed: 12/27/2022] Open
Abstract
FOXC1 is a member of Forkhead box family transcription factors. We showed that FOXC1 level was increased in melanoma cells and tissues and correlated with hypomethylation of the FOXC1 gene. Overexpression of FOXC1 promoted proliferation, migration, invasion, colony formation and growth in 3D Matrigel of melanoma cells. FOXC1 increased MST1R and activated the PI3K/AKT pathway. Also, FOXC1 expression was associated with disease progression and poor prognosis of melanoma. We suggest that FOXC1 is a potential prognostic biomarker for treating melanoma and predicting outcome of patients.
Collapse
Affiliation(s)
- Jinhua Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Department of Molecular Oncology, John Wayne Cancer Institute (JWCI) at Providence Saint John's Health Center, Santa Monica 90404, CA, USA
| | - Li Li
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Shiwei Liu
- Department of Endocrinology, Shanxi DAYI Hospital, Shanxi Medical University, Taiyuan, Shanxi 030002, China
| | - Ying Zhao
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Lin Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Guanhua Du
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
39
|
Transgenic mouse model of IgM + lymphoproliferative disease mimicking Waldenström macroglobulinemia. Blood Cancer J 2016; 6:e488. [PMID: 27813533 PMCID: PMC5148059 DOI: 10.1038/bcj.2016.95] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 09/16/2016] [Indexed: 12/19/2022] Open
Abstract
Waldenström macroglobulinemia (WM) is a low-grade incurable immunoglobulin M+ (IgM+) lymphoplasmacytic lymphoma for which a genetically engineered mouse model of de novo tumor development is lacking. On the basis of evidence that the pro-inflammatory cytokine, interleukin 6 (IL6), and the survival-enhancing oncoprotein, B cell leukemia 2 (BCL2), have critical roles in the natural history of WM, we hypothesized that the enforced expression of IL6 and BCL2 in mice unable to perform immunoglobulin class switch recombination may result in a lymphoproliferative disease that mimics WM. To evaluate this possibility, we generated compound transgenic BALB/c mice that harbored the human BCL2 and IL6 transgenes, EμSV-BCL2-22 and H2-Ld-hIL6, on the genetic background of activation-induced cytidine deaminase (AID) deficiency. We designated these mice BCL2+IL6+AID- and found that they developed-with full genetic penetrance (100% incidence) and suitably short latency (93 days median survival)-a severe IgM+ lymphoproliferative disorder that recapitulated important features of human WM. However, the BCL2+IL6+AID- model also exhibited shortcomings, such as low serum IgM levels and histopathological changes not seen in patients with WM, collectively indicating that further refinements of the model are required to achieve better correlations with disease characteristics of WM.
Collapse
|
40
|
Wang SH, Ma F, Tang ZH, Wu XC, Cai Q, Zhang MD, Weng MZ, Zhou D, Wang JD, Quan ZW. Long non-coding RNA H19 regulates FOXM1 expression by competitively binding endogenous miR-342-3p in gallbladder cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:160. [PMID: 27716361 PMCID: PMC5048611 DOI: 10.1186/s13046-016-0436-6] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/22/2016] [Indexed: 01/17/2023]
Abstract
Background Long non-coding RNA (lncRNA) H19 has been reported to involve in many kinds of human cancers and functions as an oncogene. Our previous study found that H19 was over-expressed in gallbladder cancer (GBC) and was shown to promote tumor development in GBC. However, the competing endogenous RNA (ceRNA) regulatory network involving H19 in GBC progression has not been fully elucidated. We aim to detect the role of H19 as a ceRNA in GBC. Methods and Results In this study, the expression of H19 and miR-342-3p were analyzed in 35 GBC tissues and matched normal tissues by using quantitative polymerase chain reaction (qRT-PCR). We demonstrated H19 was overexpressed and negatively correlated with miR-342-3p in GBC. By dual-luciferase reporter assays, RNA-binding protein immunoprecipitation (RIP) and RNA pull-down assays, we verified that H19 was identified as a direct target of miR-342-3p. QRT-PCR and Western-blotting assays demonstrated that H19 silencing down-regulated, whereas over-expression enhanced the expression of miR-342-3p targeting FOXM1 through competitively ‘sponging’ miR-342-3p. Furthermore, transwell invasion assays and cell cycle assays indicated that H19 knockdown inhibited both cells invasion and proliferation, but this effects was attenuated by co-transfection of siRNA-H19 and miR-342-3p inhibitor in GBC cells. In vivo, tumor volumes were decreased significantly in H19 silenced group compared to the control group, but was attenuated by co-transfection of shRNA-H19 and miR-342-3p inhibitor, which were stablely constructed through lenti-virus vector. Conclusion Our results suggest a potential ceRNA regulatory network involving H19 regulates FOXM1 expression by competitively binding endogenous miR-342-3p in GBC. This mechanism may contribute to a better understanding of GBC pathogenesis and provides potential therapeutic strategy for GBC.
Collapse
Affiliation(s)
- Shou-Hua Wang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, 200000, China
| | - Fei Ma
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiao tong University School of Medicine, Shanghai, 200092, China
| | - Zhao-Hui Tang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, 200000, China
| | - Xiao-Cai Wu
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, 200000, China
| | - Qiang Cai
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, 200000, China
| | - Ming-Di Zhang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, 200000, China
| | - Ming-Zhe Weng
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, 200000, China
| | - Di Zhou
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, 200000, China
| | - Jian-Dong Wang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, 200000, China.
| | - Zhi-Wei Quan
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, 200000, China.
| |
Collapse
|