1
|
Zhang J, Duan Y, Wu P, Chang Y, Wang Y, Hu T, Liu C, Chen X, Zong S, Chen X, Wu Y, Jin L, Lan Y, Liu X, Cheng X, Ding F, Li T, Chen X, Guo Y, Chen Y, Yang W, Zhang L, Zou Y, Cheng T, Zhu X, Zhang Y. Clonal evolution dissection reveals that a high MSI2 level promotes chemoresistance in T-cell acute lymphoblastic leukemia. Blood 2024; 143:320-335. [PMID: 37801708 DOI: 10.1182/blood.2023020490] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 10/08/2023] Open
Abstract
ABSTRACT T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive cancer with resistant clonal propagation in recurrence. We performed high-throughput droplet-based 5' single-cell RNA with paired T-cell receptor (TCR) sequencing of paired diagnosis-relapse (Dx_Rel) T-ALL samples to dissect the clonal diversities. Two leukemic evolutionary patterns, "clonal shift" and "clonal drift" were unveiled. Targeted single-cell DNA sequencing of paired Dx_Rel T-ALL samples further corroborated the existence of the 2 contrasting clonal evolution patterns, revealing that dynamic transcriptional variation might cause the mutationally static clones to evolve chemotherapy resistance. Analysis of commonly enriched drifted gene signatures showed expression of the RNA-binding protein MSI2 was significantly upregulated in the persistent TCR clonotypes at relapse. Integrated in vitro and in vivo functional studies suggested that MSI2 contributed to the proliferation of T-ALL and promoted chemotherapy resistance through the posttranscriptional regulation of MYC, pinpointing MSI2 as an informative biomarker and novel therapeutic target in T-ALL.
Collapse
Affiliation(s)
- Jingliao Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yongjuan Duan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Peng Wu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | | | - Yue Wang
- Novogene Co, Ltd, Beijing, China
| | - Tianyuan Hu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Chao Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiaoyan Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Suyu Zong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiaoli Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yangping Wu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Linlin Jin
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yang Lan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiaoming Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xuelian Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | | | - Tianyu Li
- Wuxi Children's Hospital, Jiangsu, China
| | - Xiaojuan Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Ye Guo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yumei Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wenyu Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Li Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yao Zou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiaofan Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yingchi Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
2
|
Zou D, Lv M, Chen Y, Niu T, Ma C, Shi C, Huang Z, Wu Y, Yang S, Wang Y, Wu N, Zhang Y, Ouyang G, Mu Q. Down-regulation of Musashi-2 exerts antileukemic effects on acute lymphoblastic leukemia cells and increases sensitivity to dexamethasone. Ann Hematol 2024; 103:141-151. [PMID: 37749318 DOI: 10.1007/s00277-023-05468-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 09/18/2023] [Indexed: 09/27/2023]
Abstract
Musashi-2 (MSI2), implicated in the oncogenesis and propagation of a broad array of malignancies, inclusive of certain leukemia, remains a nascent field of study within the context of acute lymphoblastic leukemia (ALL). Using lentiviral transfection, ALL cells with stable MSI2 knockdown were engineered. A suite of analytic techniques - a CCK-8 assay, flow cytometry, qRT-PCR, and western blotting - were employed to evaluate cellular proliferation, cell cycle arrest, and apoptosis and to confirm differential gene expression. The suppression of MSI2 expression yielded significant results: inhibition of cell proliferation, G0/G1 cell cycle arrest, and induced apoptosis in ALL cell lines. Furthermore, it was noted that MSI2 inhibition heightened the responsiveness of ALL cells to dexamethasone. Significantly, the depletion of MSI2 prompted the translocation of GR from the cytoplasm to the nucleus upon dexamethasone treatment, consequently leading to enhanced sensitivity. Additionally, the FOXO1/4 signaling pathway contributed to the biological effects of ALL cells evoked by MSI2 silencing. Our study offers novel insight into the inhibitory effects of MSI2 suppression on ALL cells, positing MSI2 as a promising therapeutic target in the treatment of ALL.
Collapse
Affiliation(s)
- Duobing Zou
- Laboratory of Stem Cell Transplantation, Ningbo First Hospital, Ningbo, Zhejiang, 315000, People's Republic of China
| | - Mei Lv
- Department of Hematology, Ningbo Chinese Medical Hospital, Ningbo, Zhejiang, 315000, People's Republic of China
| | - Ying Chen
- Laboratory of Stem Cell Transplantation, Ningbo First Hospital, Ningbo, Zhejiang, 315000, People's Republic of China
| | - Tingting Niu
- Laboratory of Stem Cell Transplantation, Ningbo First Hospital, Ningbo, Zhejiang, 315000, People's Republic of China
| | - Chao Ma
- Laboratory of Stem Cell Transplantation, Ningbo First Hospital, Ningbo, Zhejiang, 315000, People's Republic of China
| | - Cong Shi
- Laboratory of Stem Cell Transplantation, Ningbo First Hospital, Ningbo, Zhejiang, 315000, People's Republic of China
| | - Zhenya Huang
- Laboratory of Stem Cell Transplantation, Ningbo First Hospital, Ningbo, Zhejiang, 315000, People's Republic of China
| | - Ying Wu
- Laboratory of Stem Cell Transplantation, Ningbo First Hospital, Ningbo, Zhejiang, 315000, People's Republic of China
| | - Shujun Yang
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang, 315000, People's Republic of China
| | - Yun Wang
- Laboratory of Stem Cell Transplantation, Ningbo First Hospital, Ningbo, Zhejiang, 315000, People's Republic of China
| | - Ningning Wu
- Laboratory of Stem Cell Transplantation, Ningbo First Hospital, Ningbo, Zhejiang, 315000, People's Republic of China
| | - Yi Zhang
- Laboratory of Stem Cell Transplantation, Ningbo First Hospital, Ningbo, Zhejiang, 315000, People's Republic of China
| | - Guifang Ouyang
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang, 315000, People's Republic of China.
| | - Qitian Mu
- Laboratory of Stem Cell Transplantation, Ningbo First Hospital, Ningbo, Zhejiang, 315000, People's Republic of China.
| |
Collapse
|
3
|
Álvarez-Zúñiga CD, Garza-Veloz I, Martínez-Rendón J, Ureño-Segura M, Delgado-Enciso I, Martinez-Fierro ML. Circulating Biomarkers Associated with the Diagnosis and Prognosis of B-Cell Progenitor Acute Lymphoblastic Leukemia. Cancers (Basel) 2023; 15:4186. [PMID: 37627214 PMCID: PMC10453581 DOI: 10.3390/cancers15164186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a hematological disease characterized by the dysfunction of the hematopoietic system that leads to arrest at a specific stage of stem cells development, suppressing the average production of cellular hematologic components. BCP-ALL is a neoplasm of the B-cell lineage progenitor. BCP-ALL is caused and perpetuated by several mechanisms that provide the disease with its tumor potential and genetic and cytological characteristics. These pathological features are used for diagnosis and the prognostication of BCP-ALL. However, most of these paraclinical tools can only be obtained by bone marrow aspiration, which, as it is an invasive study, can delay the diagnosis and follow-up of the disease, in addition to the anesthetic risk it entails for pediatric patients. For this reason, it is crucial to find noninvasive and accessible ways to supply information concerning diagnosis, prognosis, and the monitoring of the disease, such as circulating biomarkers. In oncology, a biomarker is any measurable indicator that demonstrates the presence of malignancy, tumoral behavior, prognosis, or responses to treatments. This review summarizes circulating molecules associated with BCP-ALL with potential diagnostic value, classificatory capacity during monitoring specific clinic features of the disease, and/or capacity to identify each BCP-ALL stage regarding its evolution and outcome of the patients with BCP-ALL. In the same way, we provide and classify biomarkers that may be used in further studies focused on clinical approaches or therapeutic target identification for BCP-ALL.
Collapse
Affiliation(s)
- Claudia Daniela Álvarez-Zúñiga
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y C.S, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico; (C.D.Á.-Z.); (I.G.-V.); (J.M.-R.)
| | - Idalia Garza-Veloz
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y C.S, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico; (C.D.Á.-Z.); (I.G.-V.); (J.M.-R.)
| | - Jacqueline Martínez-Rendón
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y C.S, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico; (C.D.Á.-Z.); (I.G.-V.); (J.M.-R.)
| | - Misael Ureño-Segura
- Hematology Service, Hospital General Zacatecas “Luz González Cosío”, Servicios de Salud de Zacatecas, Zacatecas 98160, Mexico;
| | - Iván Delgado-Enciso
- Cancerology State Institute, Colima State Health Services, Colima 28085, Mexico;
- School of Medicine, University of Colima, Colima 28040, Mexico
| | - Margarita L. Martinez-Fierro
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y C.S, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico; (C.D.Á.-Z.); (I.G.-V.); (J.M.-R.)
| |
Collapse
|
4
|
Wu W, Li J, Dong D, Dou F, Lin Y, Yang X, Zhou Y, Xie J. Prognostic value of MSI2 expression in human malignancies: A PRISMA-compliant meta-analysis. Medicine (Baltimore) 2022; 101:e32064. [PMID: 36596017 PMCID: PMC9803470 DOI: 10.1097/md.0000000000032064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The prognostic value of Musashi-2 (MSI2) in human malignancies remains controversial. We thus conducted this meta-analysis to evaluate the association between MSI2 expression and prognosis of patients with malignancies. MATERIALS AND METHODS We searched EMBASE, PubMed and Web of Science up to June 2021 for eligible studies. Hazard ratios (HRs) with 95% confidence intervals (CIs) were estimated to assess the prognostic value of MSI2 expression. Odds ratios (ORs) with 95% CIs were calculated to evaluate the association between MSI2 expression and clinicopathological traits. RESULTS Sixteen studies involving 2203 patients were finally included in this meta-analysis. We found that high MSI2 expression might predict unfavorable OS (HR = 1.85, 95% CI: 1.62-2.10, P < .0001) and DFS/RFS (HR = 2.19, 95% CI: 1.87-2.57, P < .0001). Besides, the pooled results indicated that increased MSI2 expression correlated with large tumor size, poor tumor differentiation, positive lymph node metastasis and advanced tumor stage. CONCLUSIONS Taken together, our data implies that MSI2 overexpression is related to poor survival outcomes in patients with malignancy. Therefore, MSI2 may serve as a novel prognostic biomarker and therapeutic target of malignancies. However, large-scale prospective and homogeneous investigations should be conducted in the future to further validate our findings.
Collapse
Affiliation(s)
- Wei Wu
- Anorectum Surgical Department, YunNan Provimcial Hospital of Traditional Chinese Medicine, YunNan, China
- Department of Gastrointestinal Surgery, 3201 Hospital of Xi’an Jiao Tong University Health Science Center, Hanzhong, Shaanxi, China
- *Correspondence: Wei Wu, Anorectum Surgical Department, YunNan Provimcial Hospital of Traditional Chinese Medicine, YunNan, China; Department of Gastrointestinal Surgery, 3201 Hospital of Xi’an Jiao Tong University Health Science Center, Hanzhong 723000, Shaanxi, China and Jun Xie, Anorectum Surgical Department, YunNan Provimcial Hospital of Traditongnal Chinese Medicine, YunNan, China (e-mail: and )
| | - Jialin Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Dejia Dong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Fafu Dou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yong Lin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaoye Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yan Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jun Xie
- Anorectum Surgical Department, YunNan Provimcial Hospital of Traditional Chinese Medicine, YunNan, China
- *Correspondence: Wei Wu, Anorectum Surgical Department, YunNan Provimcial Hospital of Traditional Chinese Medicine, YunNan, China; Department of Gastrointestinal Surgery, 3201 Hospital of Xi’an Jiao Tong University Health Science Center, Hanzhong 723000, Shaanxi, China and Jun Xie, Anorectum Surgical Department, YunNan Provimcial Hospital of Traditongnal Chinese Medicine, YunNan, China (e-mail: and )
| |
Collapse
|
5
|
Jiang L, Xue S, Xu J, Fu X, Wei J, Zhang C. Prognostic value of Musashi 2 (MSI2) in cancer patients: A systematic review and meta-analysis. Front Oncol 2022; 12:969632. [PMID: 36530989 PMCID: PMC9751961 DOI: 10.3389/fonc.2022.969632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 11/02/2022] [Indexed: 11/10/2023] Open
Abstract
Musashi 2 (MSI2) is an RNA-binding protein that regulates mRNA translation of numerous intracellular targets and plays an important role in the development of cancer. However, the prognostic value of MSI2 in various cancers remains controversial. Herein, we conducted this meta-analysis including 21 studies with 2640 patients searched from PubMed, Web of Science, EMBASE, Chinese National Knowledge Infrastructure databases, and WanFang databases to accurately assess the prognostic significance of MSI2 in various cancers. Our results indicated that high MSI2 expression was significantly related to poor overall survival (HR = 1.84, 95% CI: 1.66-2.05, P < 0.001) and disease-free survival (HR = 1.73, 95% CI: 1.35-2.22, P < 0.001). In addition, MSI2 positive expression was associated with certain phenotypes of tumor aggressiveness, such as clinical stage, depth of invasion, lymph node metastasis, liver metastasis and tumor size. In conclusion, elevated MSI2 expression is closely correlated with poor prognosis in various cancers, and may serve as a potential molecular target for cancer patients.
Collapse
Affiliation(s)
- Lin Jiang
- Department of Anesthesiology, Taizhou People’s Hospital, Affiliated to Nanjing Medical University, Taizhou, Jiangsu, China
| | - Shanshan Xue
- Department of Clinical Laboratory, Taizhou People’s Hospital, Affiliated to Nanjing Medical University, Taizhou, Jiangsu, China
| | - Jie Xu
- The Center for Translational Medicine, Taizhou People’s Hospital, Affiliated to Nanjing Medical University, Taizhou, Jiangsu, China
| | - Xiaoyang Fu
- The Center for Translational Medicine, Taizhou People’s Hospital, Affiliated to Nanjing Medical University, Taizhou, Jiangsu, China
| | - Jing Wei
- Department of Obstetrics and Gynecology, Taizhou People’s Hospital, Affiliated to Nanjing Medical University, Taizhou, Jiangsu, China
| | - Chuanmeng Zhang
- The Center for Translational Medicine, Taizhou People’s Hospital, Affiliated to Nanjing Medical University, Taizhou, Jiangsu, China
| |
Collapse
|
6
|
Luo DD, Zhao F. KLF4 suppresses the proliferation and metastasis of NSCLC cells via inhibition of MSI2 and regulation of the JAK/STAT3 signaling pathway. Transl Oncol 2022; 22:101396. [PMID: 35580385 PMCID: PMC9117691 DOI: 10.1016/j.tranon.2022.101396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 11/18/2022] Open
Abstract
KLF4 is downregulated, while MSI2 is upregulated in NSCLC tissues. Overexpression of KLF4 suppresses NSCLC cell migration and invasion. Knockdown of MSI2 suppresses the migration and invasion of NSCLC cells. KLF4 inhibits the transcription of MSI2 in NSCLC cells. KLF4 suppressed the invasion and migration of NSCLC cells via inhibition of JAK2/STAT3 signalling pathway.
Background Non-small cell lung cancer (NSCLC) remains an aggresive tumor with poor survival rates. Krüppel-like factor 4 (KLF4) is known to be involved in progression of NSCLC; however, the detailed mechanism by which KLF4 regulates the progression of NSCLC remains unclear. Methods In order to investigate the function of KLF4 in NSCLC, cell proliferation was measured by MTT and colony formation assays. The migration and invasion of NSCLC cells were detected via wound healing and Transwell assays, respectively. Then, the interaction between KLF4 and MSI2 was confirmed using a dual-luciferase reporter assay, and the mechanism by which KLF4 regulates the tumorigenesis of NSCLC was assessed by RT-qPCR and Western blotting. Results The results showed that KLF4 was downregulated, while MSI2 was upregulated in NSCLC. Additionally, KLF4 could inhibit transcription of MSI2, and overexpression of KLF4 or knockdown of MSI2 could inhibit the proliferation, migration and invasion of NSCLC cells. Moreover, KLF4 could inhibit JAK2/STAT3 signalling pathway. Conclusions In conclusion, KLF4 significantly inhibited the proliferation, invasion and migration of NSCLC cells via inactivation of MSI2/JAK2/STAT3 signalling pathway. Thereby, our finding might shed new lights on exploring the new strategies against NSCLC.
Collapse
|
7
|
Gladbach YS, Sklarz LM, Roolf C, Beck J, Schütz E, Fuellen G, Junghanss C, Murua Escobar H, Hamed M. Molecular Characterization of the Response to Conventional Chemotherapeutics in Pro-B-ALL Cell Lines in Terms of Tumor Relapse. Genes (Basel) 2022; 13:genes13071240. [PMID: 35886023 PMCID: PMC9316692 DOI: 10.3390/genes13071240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/26/2022] [Accepted: 06/29/2022] [Indexed: 11/29/2022] Open
Abstract
Little is known about optimally applying chemotherapeutic agents in a specific temporal sequence to rapidly reduce the tumor load and to improve therapeutic efficacy. The clinical optimization of drug efficacy while reducing side effects is still restricted due to an incomplete understanding of the mode of action and related tumor relapse mechanisms on the molecular level. The molecular characterization of transcriptomic drug signatures can help to identify the affected pathways, downstream regulated genes and regulatory interactions related to tumor relapse in response to drug application. We tried to outline the dynamic regulatory reprogramming leading to tumor relapse in relapsed MLL-rearranged pro-B-cell acute lymphoblastic leukemia (B-ALL) cells in response to two first-line treatments: dexamethasone (Dexa) and cytarabine (AraC). We performed an integrative molecular analysis of whole transcriptome profiles of each treatment, specifically considering public knowledge of miRNA regulation via a network-based approach to unravel key driver genes and miRNAs that may control the relapse mechanisms accompanying each treatment. Our results gave hints to the crucial regulatory roles of genes leading to Dexa-resistance and related miRNAs linked to chemosensitivity. These genes and miRNAs should be further investigated in preclinical models to obtain more hints about relapse processes.
Collapse
Affiliation(s)
- Yvonne Saara Gladbach
- Institute for Biostatistics and Informatics in Medicine and Ageing Research (IBIMA), Rostock University Medical Center, 18057 Rostock, Germany; (Y.S.G.); (G.F.)
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Lisa-Madeleine Sklarz
- Clinic III—Hematology, Oncology, Palliative Medicine, Center for Internal Medicine, Rostock University Medical Center, 18057 Rostock, Germany; (L.-M.S.); (C.R.); (C.J.); (H.M.E.)
| | - Catrin Roolf
- Clinic III—Hematology, Oncology, Palliative Medicine, Center for Internal Medicine, Rostock University Medical Center, 18057 Rostock, Germany; (L.-M.S.); (C.R.); (C.J.); (H.M.E.)
| | - Julia Beck
- Chronix Biomedical GmbH, 37073 Göttingen, Germany; (J.B.); (E.S.)
| | - Ekkehard Schütz
- Chronix Biomedical GmbH, 37073 Göttingen, Germany; (J.B.); (E.S.)
| | - Georg Fuellen
- Institute for Biostatistics and Informatics in Medicine and Ageing Research (IBIMA), Rostock University Medical Center, 18057 Rostock, Germany; (Y.S.G.); (G.F.)
| | - Christian Junghanss
- Clinic III—Hematology, Oncology, Palliative Medicine, Center for Internal Medicine, Rostock University Medical Center, 18057 Rostock, Germany; (L.-M.S.); (C.R.); (C.J.); (H.M.E.)
| | - Hugo Murua Escobar
- Clinic III—Hematology, Oncology, Palliative Medicine, Center for Internal Medicine, Rostock University Medical Center, 18057 Rostock, Germany; (L.-M.S.); (C.R.); (C.J.); (H.M.E.)
- Comprehensive Cancer Center Mecklenburg-Vorpommern (CCC-MV), Campus Rostock, Rostock University Medical Center, 18057 Rostock, Germany
| | - Mohamed Hamed
- Institute for Biostatistics and Informatics in Medicine and Ageing Research (IBIMA), Rostock University Medical Center, 18057 Rostock, Germany; (Y.S.G.); (G.F.)
- Correspondence:
| |
Collapse
|
8
|
Veronez LC, das Chagas PF, Corrêa CAP, Baroni M, da Silva KR, Nagano LF, Borges KS, Queiroz RGP, Tone LG, Scrideli CA. MSI2 expression in adrenocortical carcinoma: Association with unfavorable prognosis and correlation with steroid and immune-related pathways. J Cell Biochem 2021; 122:1925-1935. [PMID: 34581457 DOI: 10.1002/jcb.30153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/23/2021] [Accepted: 09/09/2021] [Indexed: 11/06/2022]
Abstract
Adrenocortical carcinoma (ACC) is a rare, but highly aggressive cancer of the adrenal cortex with a generally poor prognosis. Despite being rare, completely resected ACCs present a high risk of recurrence. Musashi-2 (MSI2) has recently been recognized as a potential prognostic biomarker and therapeutic target in many cancers. However, no studies have evaluated the clinical significance of MSI2 expression in ACC. Here, we addressed MSI2 expression and its association with ACC prognosis and clinicopathological parameters. MSI2 expression was analyzed in TCGA, GSE12368, GSE33371, and GSE49278 ACC datasets; and its correlation with other genes and immune cell infiltration were investigated by using the R2: Genomics Analysis and Visualization Platform and TIMER databases, respectively. Enrichment analysis was performed with the DAVID Functional Annotation Tool. Kaplan-Meier curves, log-rank tests, and Cox regression analyses were used to explore the prognostic role of MSI2 in ACC. Our findings demonstrated the potential value of MSI2 overexpression as an independent predictor of poor prognosis in patients with completely resected ACC (hazard ratio 6.715, 95% confidence interval 1.266 - 35.620, p =.025). In addition, MSI2 overexpression was associated with characteristics of unfavorable prognosis, such as cortisol excess (p = .002), recurrence (p =.003), and death (p =.015); positively correlated with genes related to steroid biosynthesis (p < .05); and negatively correlated with immune-related pathways (p < .05). Our findings demonstrate that MSI2 has value as a prognostic marker for completely resected ACC and reinforce the investigation of its role as a possible therapeutic target for patients with ACC.
Collapse
Affiliation(s)
- Luciana C Veronez
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Pablo F das Chagas
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Carolina A P Corrêa
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Mirella Baroni
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Keteryne R da Silva
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Luis F Nagano
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Kleiton S Borges
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Rosane G P Queiroz
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Luiz G Tone
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Carlos A Scrideli
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
9
|
Heyes E, Schmidt L, Manhart G, Eder T, Proietti L, Grebien F. Identification of gene targets of mutant C/EBPα reveals a critical role for MSI2 in CEBPA-mutated AML. Leukemia 2021; 35:2526-2538. [PMID: 33623142 PMCID: PMC7611617 DOI: 10.1038/s41375-021-01169-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 01/12/2021] [Accepted: 01/28/2021] [Indexed: 01/31/2023]
Abstract
Mutations in the gene encoding the transcription factor CCAAT/enhancer-binding protein alpha (C/EBPα) occur in 10-15% of acute myeloid leukemia (AML). Frameshifts in the CEBPA N-terminus resulting in exclusive expression of a truncated p30 isoform represent the most prevalent type of CEBPA mutations in AML. C/EBPα p30 interacts with the epigenetic machinery, but it is incompletely understood how p30-induced changes cause leukemogenesis. We hypothesized that critical effector genes in CEBPA-mutated AML are dependent on p30-mediated dysregulation of the epigenome. We mapped p30-associated regulatory elements (REs) by ATAC-seq and ChIP-seq in a myeloid progenitor cell model for p30-driven AML that enables inducible RNAi-mediated knockdown of p30. Concomitant p30-dependent changes in gene expression were measured by RNA-seq. Integrative analysis identified 117 p30-dependent REs associated with 33 strongly down-regulated genes upon p30-knockdown. CRISPR/Cas9-mediated mutational disruption of these genes revealed the RNA-binding protein MSI2 as a critical p30-target. MSI2 knockout in p30-driven murine AML cells and in the CEBPA-mutated human AML cell line KO-52 caused proliferation arrest and terminal myeloid differentiation, and delayed leukemia onset in vivo. In summary, this work presents a comprehensive dataset of p30-dependent effects on epigenetic regulation and gene expression and identifies MSI2 as an effector of the C/EBPα p30 oncoprotein.
Collapse
Affiliation(s)
- Elizabeth Heyes
- University of Veterinary Medicine, Institute of Medical Biochemistry, Vienna, Austria
| | - Luisa Schmidt
- University of Veterinary Medicine, Institute of Medical Biochemistry, Vienna, Austria
| | - Gabriele Manhart
- University of Veterinary Medicine, Institute of Medical Biochemistry, Vienna, Austria
| | - Thomas Eder
- University of Veterinary Medicine, Institute of Medical Biochemistry, Vienna, Austria
| | - Ludovica Proietti
- University of Veterinary Medicine, Institute of Medical Biochemistry, Vienna, Austria
| | - Florian Grebien
- University of Veterinary Medicine, Institute of Medical Biochemistry, Vienna, Austria.
| |
Collapse
|
10
|
Kharin L, Bychkov I, Karnaukhov N, Voloshin M, Fazliyeva R, Deneka A, Frantsiyants E, Kit O, Golemis E, Boumber Y. Prognostic role and biologic features of Musashi-2 expression in colon polyps and during colorectal cancer progression. PLoS One 2021; 16:e0252132. [PMID: 34237057 PMCID: PMC8266110 DOI: 10.1371/journal.pone.0252132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 05/10/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The RNA-binding protein Musashi-2 (MSI2) controls the translation of proteins that support stem cell identity and lineage determination and is associated with progression in some cancers. We assessed MSI2 as potential clinical biomarker in colorectal cancer (CRC) and tubulovillous adenoma (TA) of colon mucosa. METHODS We assessed 125 patients, of whom 20 had polyps of the colon (TAs), and 105 had CRC. Among 105 patients with CRC, 45 had stages I-III; among metastatic CRC (mCRC) patients, 31 had synchronous and 29 metachronous liver metastases. We used immunohistochemistry to measure MSI2 expression in matching specimens of normal tissue versus TAs, primary CRC tumors, and metastases, correlating expression to clinical outcomes. We analyzed the biological effects of depleting MSI2 expression in human CRC cells. RESULTS MSI2 expression was significantly elevated in polyps versus primary tissue, and further significantly elevated in primary tumors and metastases. MSI2 expression correlated with decreased progression free survival (PFS) and overall survival (OS), higher tumor grade, and right-side localization (p = 0.004) of tumors. In metastases, high MSI2 expression correlated with E-cadherin expression. Knockdown of MSI2 in CRC cells suppressed proliferation, survival and clonogenic capacity, and decreased expression of TGFβ1, E-cadherin, and ZO1. CONCLUSION Elevated expression of MSI2 is associated with pre-cancerous TAs in the colonic mucosa, suggesting it is an early event in transformation. MSI2 expression is further elevated during CRC progression, and associated with poor prognosis. Depletion of MSI2 reduces CRC cell growth. These data imply a causative role of MSI2 overexpression at multiple stages of CRC formation and progression.
Collapse
Affiliation(s)
- Leonid Kharin
- National Medical Research Center of Oncology, Rostov-on-Don, Russian Federation
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Igor Bychkov
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Nikolay Karnaukhov
- National Medical Research Center of Oncology, Rostov-on-Don, Russian Federation
| | - Mark Voloshin
- Rostov State Medical University, Rostov-on-Don, Russian Federation
| | - Rushaniya Fazliyeva
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Alexander Deneka
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Elena Frantsiyants
- National Medical Research Center of Oncology, Rostov-on-Don, Russian Federation
| | - Oleg Kit
- National Medical Research Center of Oncology, Rostov-on-Don, Russian Federation
| | - Erica Golemis
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Yanis Boumber
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
- Kazan Federal University, Kazan, Russian Federation
- Department of Hematology/Oncology, Section of Thoracic Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
11
|
Sun J, Sheng W, Ma Y, Dong M. Potential Role of Musashi-2 RNA-Binding Protein in Cancer EMT. Onco Targets Ther 2021; 14:1969-1980. [PMID: 33762829 PMCID: PMC7982713 DOI: 10.2147/ott.s298438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/22/2021] [Indexed: 12/20/2022] Open
Abstract
Local invasion and distant metastasis are the key hallmarks in the aggressive progression of malignant tumors, including the ability of cancer cells to detach from the extracellular matrix overcome apoptosis, and disseminate into distant sites. It is generally believed that this malignant behavior is stimulated by epithelial-mesenchymal transition (EMT). Musashi (MSI) RNA-binding proteins, belonging to the evolutionarily conserved RNA-binding proteins (RBP) family, were originally discovered to regulate asymmetric cell division during embryonic development. Recently, Musashi-2 (MSI2), as a key member of MSI family, has been prevalently reported to be tightly associated with the advanced clinical stage of several cancers. Multiple oncogenic signaling pathways mediated by MSI2 play vital roles in EMT. Here, we systematically reviewed the detailed role and signal networks of MSI2 in regulating cancer development, especially in EMT signal transduction, involving EGF, TGF-β, Notch, and Wnt pathways.
Collapse
Affiliation(s)
- Jian Sun
- Department of Gastrointestinal Surgery, The First Hospital, China Medical University, Shenyang, 110001, People's Republic of China
| | - Weiwei Sheng
- Department of Gastrointestinal Surgery, The First Hospital, China Medical University, Shenyang, 110001, People's Republic of China
| | - Yuteng Ma
- Department of Gastrointestinal Surgery, The First Hospital, China Medical University, Shenyang, 110001, People's Republic of China
| | - Ming Dong
- Department of Gastrointestinal Surgery, The First Hospital, China Medical University, Shenyang, 110001, People's Republic of China
| |
Collapse
|
12
|
Topchu I, Karnaukhov N, Mazitova A, Yugai V, Voloshin M, Tikhomirova M, Kit O, Frantsiyants E, Kharin L, Airapetova T, Ratner E, Sabirov A, Abramova Z, Serebriiskii I, Boumber Y, Deneka A. Musashi 2 (MSI2) expression as an independent prognostic biomarker in non-small cell lung cancer (NSCLC). J Thorac Dis 2021; 13:1370-1379. [PMID: 33841930 PMCID: PMC8024834 DOI: 10.21037/jtd-20-2787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Musashi-2 (MSI2) is a member of RNA-binding protein family that regulates mRNA translation of numerous intracellular targets and influences maintenance of stem cell identity. This study assessed MSI2 as a potential clinical biomarker in non-small cell lung cancer (NSCLC). Methods The current study included 40 patients with NSCLC, of whom one presented with stage 1, 14 presented with stage II, 15 presented with stage III, and 10 patients had stage IV. All patients received standard of care treatments. All patient samples were obtained before treatment started. We used immunohistochemical (IHC) approach to measure MSI2 protein expression in matching specimens of normal lung versus tumor tissues, and primary versus metastatic tumors, followed by correlative analysis in relation to clinical outcomes. In parallel, clinical correlative analysis of MSI2 mRNA expression was performed in silico using publicly available datasets (TCGA/ICGC and KM plots). Results MSI2 protein expression in patient samples was significantly elevated in NSCLC primary tumors versus normal lung tissue (P=0.03). MSI2 elevated expression positively correlated with a decreased progression free survival (PFS) (P=0.026) combined for all stages and with overall survival (OS) at stage IV (P=0.013). Elevated MSI2 expression on RNA level was confirmed in primary tumor versus normal tissue samples in TCGA dataset (P<0.0001), and positively correlated with decreased OS (P=0.02). No correlation was observed between MSI2 expression and age, sex, smoking, and treatment type. Conclusions Elevated MSI2 expression in primary NSCLC tumors is associated with poor prognosis and can be used as a novel potential prognostic biomarker in NSCLC patients. Future studies in an extended patient cohort are warranted.
Collapse
Affiliation(s)
- Iuliia Topchu
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA.,Kazan Federal University, Kazan, Russian Federation
| | - Nikolai Karnaukhov
- National Medical Research Center of Oncology, Rostov-on-Don, Russia, Russian Federation
| | - Alexandra Mazitova
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA.,Kazan Federal University, Kazan, Russian Federation
| | | | - Mark Voloshin
- National Medical Research Center of Oncology, Rostov-on-Don, Russia, Russian Federation
| | | | - Oleg Kit
- National Medical Research Center of Oncology, Rostov-on-Don, Russia, Russian Federation
| | - Elena Frantsiyants
- National Medical Research Center of Oncology, Rostov-on-Don, Russia, Russian Federation
| | - Leonid Kharin
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA.,National Medical Research Center of Oncology, Rostov-on-Don, Russia, Russian Federation
| | - Tamara Airapetova
- National Medical Research Center of Oncology, Rostov-on-Don, Russia, Russian Federation
| | - Ekaterina Ratner
- Tatarstan Regional Clinical Cancer Center, Kazan, Russian Federation
| | - Alexey Sabirov
- Tatarstan Regional Clinical Cancer Center, Kazan, Russian Federation
| | | | - Iliya Serebriiskii
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA.,Kazan Federal University, Kazan, Russian Federation
| | - Yanis Boumber
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA.,Kazan Federal University, Kazan, Russian Federation.,Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Alexander Deneka
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA.,Kazan Federal University, Kazan, Russian Federation
| |
Collapse
|
13
|
Schwartz JR, Ma J, Kamens J, Westover T, Walsh MP, Brady SW, Robert Michael J, Chen X, Montefiori L, Song G, Wu G, Wu H, Branstetter C, Hiltenbrand R, Walsh MF, Nichols KE, Maciaszek JL, Liu Y, Kumar P, Easton J, Newman S, Rubnitz JE, Mullighan CG, Pounds S, Zhang J, Gruber T, Ma X, Klco JM. The acquisition of molecular drivers in pediatric therapy-related myeloid neoplasms. Nat Commun 2021; 12:985. [PMID: 33579957 PMCID: PMC7880998 DOI: 10.1038/s41467-021-21255-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 01/15/2021] [Indexed: 12/21/2022] Open
Abstract
Pediatric therapy-related myeloid neoplasms (tMN) occur in children after exposure to cytotoxic therapy and have a dismal prognosis. The somatic and germline genomic alterations that drive these myeloid neoplasms in children and how they arise have yet to be comprehensively described. We use whole exome, whole genome, and/or RNA sequencing to characterize the genomic profile of 84 pediatric tMN cases (tMDS: n = 28, tAML: n = 56). Our data show that Ras/MAPK pathway mutations, alterations in RUNX1 or TP53, and KMT2A rearrangements are frequent somatic drivers, and we identify cases with aberrant MECOM expression secondary to enhancer hijacking. Unlike adults with tMN, we find no evidence of pre-existing minor tMN clones (including those with TP53 mutations), but rather the majority of cases are unrelated clones arising as a consequence of cytotoxic therapy. These studies also uncover rare cases of lineage switch disease rather than true secondary neoplasms.
Collapse
Affiliation(s)
- Jason R Schwartz
- Vanderbilt University Medical Center, Department of Pediatrics, Nashville, TN, US
| | - Jing Ma
- St. Jude Children's Research Hospital, Department of Pathology, Memphis, TN, US
| | - Jennifer Kamens
- Stanford University School of Medicine, Department of Pediatrics, Stanford, CA, US
| | - Tamara Westover
- St. Jude Children's Research Hospital, Department of Pathology, Memphis, TN, US
| | - Michael P Walsh
- St. Jude Children's Research Hospital, Department of Pathology, Memphis, TN, US
| | - Samuel W Brady
- St. Jude Children's Research Hospital, Department of Computational Biology, Memphis, TN, US
| | - J Robert Michael
- St. Jude Children's Research Hospital, Department of Computational Biology, Memphis, TN, US
| | - Xiaolong Chen
- St. Jude Children's Research Hospital, Department of Computational Biology, Memphis, TN, US
| | - Lindsey Montefiori
- St. Jude Children's Research Hospital, Department of Pathology, Memphis, TN, US
| | - Guangchun Song
- St. Jude Children's Research Hospital, Department of Pathology, Memphis, TN, US
| | - Gang Wu
- St. Jude Children's Research Hospital, Department of Computational Biology, Memphis, TN, US
| | - Huiyun Wu
- St. Jude Children's Research Hospital, Department of Biostatistics, Memphis, TN, US
| | - Cristyn Branstetter
- Arkansas Children's Northwest Hospital, Department of Hematology/Oncology, Springdale, AR, US
| | - Ryan Hiltenbrand
- St. Jude Children's Research Hospital, Department of Pathology, Memphis, TN, US
| | - Michael F Walsh
- Memorial Sloan Kettering Cancer Center, Department of Pediatrics, New York, NY, US
| | - Kim E Nichols
- St. Jude Children's Research Hospital, Department of Oncology, Memphis, TN, US
| | - Jamie L Maciaszek
- St. Jude Children's Research Hospital, Department of Oncology, Memphis, TN, US
| | - Yanling Liu
- St. Jude Children's Research Hospital, Department of Computational Biology, Memphis, TN, US
| | - Priyadarshini Kumar
- St. Jude Children's Research Hospital, Department of Pathology, Memphis, TN, US
| | - John Easton
- St. Jude Children's Research Hospital, Department of Computational Biology, Memphis, TN, US
| | - Scott Newman
- St. Jude Children's Research Hospital, Department of Computational Biology, Memphis, TN, US
| | - Jeffrey E Rubnitz
- St. Jude Children's Research Hospital, Department of Oncology, Memphis, TN, US
| | - Charles G Mullighan
- St. Jude Children's Research Hospital, Department of Pathology, Memphis, TN, US
| | - Stanley Pounds
- St. Jude Children's Research Hospital, Department of Biostatistics, Memphis, TN, US
| | - Jinghui Zhang
- St. Jude Children's Research Hospital, Department of Computational Biology, Memphis, TN, US
| | - Tanja Gruber
- Stanford University School of Medicine, Department of Pediatrics, Stanford, CA, US.
- Stanford University School of Medicine, Stanford Cancer Institute, Stanford, CA, US.
| | - Xiaotu Ma
- St. Jude Children's Research Hospital, Department of Computational Biology, Memphis, TN, US.
| | - Jeffery M Klco
- St. Jude Children's Research Hospital, Department of Pathology, Memphis, TN, US.
| |
Collapse
|
14
|
Bitaraf A, Razmara E, Bakhshinejad B, Yousefi H, Vatanmakanian M, Garshasbi M, Cho WC, Babashah S. The oncogenic and tumor suppressive roles of RNA-binding proteins in human cancers. J Cell Physiol 2021; 236:6200-6224. [PMID: 33559213 DOI: 10.1002/jcp.30311] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 01/14/2021] [Accepted: 01/22/2021] [Indexed: 12/17/2022]
Abstract
Posttranscriptional regulation is a mechanism for the cells to control gene regulation at the RNA level. In this process, RNA-binding proteins (RBPs) play central roles and orchestrate the function of RNA molecules in multiple steps. Accumulating evidence has shown that the aberrant regulation of RBPs makes contributions to the initiation and progression of tumorigenesis via numerous mechanisms such as genetic changes, epigenetic alterations, and noncoding RNA-mediated regulations. In this article, we review the effects caused by RBPs and their functional diversity in the malignant transformation of cancer cells that occurs through the involvement of these proteins in various stages of RNA regulation including alternative splicing, stability, polyadenylation, localization, and translation. Besides this, we review the various interactions between RBPs and other crucial posttranscriptional regulators such as microRNAs and long noncoding RNAs in the pathogenesis of cancer. Finally, we discuss the potential approaches for targeting RBPs in human cancers.
Collapse
Affiliation(s)
- Amirreza Bitaraf
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ehsan Razmara
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Babak Bakhshinejad
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, LSUHSC School of Medicine, New Orleans, Louisiana, USA
| | - Mousa Vatanmakanian
- Department of Biochemistry and Molecular Biology, LSUHSC School of Medicine, New Orleans, Louisiana, USA
| | - Masoud Garshasbi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
15
|
RNA-Binding Proteins in Acute Leukemias. Int J Mol Sci 2020; 21:ijms21103409. [PMID: 32408494 PMCID: PMC7279408 DOI: 10.3390/ijms21103409] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/07/2020] [Accepted: 05/10/2020] [Indexed: 12/12/2022] Open
Abstract
Acute leukemias are genetic diseases caused by translocations or mutations, which dysregulate hematopoiesis towards malignant transformation. However, the molecular mode of action is highly versatile and ranges from direct transcriptional to post-transcriptional control, which includes RNA-binding proteins (RBPs) as crucial regulators of cell fate. RBPs coordinate RNA dynamics, including subcellular localization, translational efficiency and metabolism, by binding to their target messenger RNAs (mRNAs), thereby controlling the expression of the encoded proteins. In view of the growing interest in these regulators, this review summarizes recent research regarding the most influential RBPs relevant in acute leukemias in particular. The reported RBPs, either dysregulated or as components of fusion proteins, are described with respect to their functional domains, the pathways they affect, and clinical aspects associated with their dysregulation or altered functions.
Collapse
|
16
|
Chagas PF, Baroni M, Brassesco MS, Tone LG. Interplay between the RNA binding‐protein Musashi and developmental signaling pathways. J Gene Med 2020; 22:e3136. [DOI: 10.1002/jgm.3136] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/19/2019] [Accepted: 10/20/2019] [Indexed: 12/17/2022] Open
Affiliation(s)
- Pablo Ferreira Chagas
- Department of GeneticsRibeirão Preto Medical School, University of São Paulo Ribeirão Preto São Paulo Brazil
| | - Mirella Baroni
- Department of GeneticsRibeirão Preto Medical School, University of São Paulo Ribeirão Preto São Paulo Brazil
| | - María Sol Brassesco
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão PretoUniversity of São Paulo Brazil
| | - Luiz Gonzaga Tone
- Department of GeneticsRibeirão Preto Medical School, University of São Paulo Ribeirão Preto São Paulo Brazil
- Department of PediatricsRibeirão Preto Medical School São Paulo
| |
Collapse
|
17
|
Lan L, Xing M, Kashipathy M, Douglas J, Gao P, Battaile K, Hanzlik R, Lovell S, Xu L. Crystal and solution structures of human oncoprotein Musashi-2 N-terminal RNA recognition motif 1. Proteins 2019; 88:573-583. [PMID: 31603583 PMCID: PMC7079100 DOI: 10.1002/prot.25836] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 09/16/2019] [Accepted: 09/28/2019] [Indexed: 01/03/2023]
Abstract
Musashi‐2 (MSI2) belongs to Musashi family of RNA binding proteins (RBP). Like Musashi‐1 (MSI1), it is overexpressed in a variety of cancers and is a promising therapeutic target. Both MSI proteins contain two N‐terminal RNA recognition motifs and play roles in posttranscriptional regulation of target mRNAs. Previously, we have identified several inhibitors of MSI1, all of which bind to MSI2 as well. In order to design MSI2‐specific inhibitors and compare the differences of binding mode of the inhibitors, we set out to solve the structure of MSI2‐RRM1, the key motif that is responsible for the binding. Here, we report the crystal structure and the first NMR solution structure of MSI2‐RRM1, and compare these to the structures of MSI1‐RBD1 and other RBPs. A high degree of structural similarity was observed between the crystal and solution NMR structures. MSI2‐RRM1 shows a highly similar overall folding topology to MSI1‐RBD1 and other RBPs. The structural information of MSI2‐RRM1 will be helpful for understanding MSI2‐RNA interaction and for guiding rational drug design of MSI2‐specific inhibitors.
Collapse
Affiliation(s)
- Lan Lan
- Department of Molecular Biosciences, The University of Kansas, Lawrence, Kansas
| | - Minli Xing
- Bio-NMR Core Facility, NIH COBRE in Protein Structure and Function, The University of Kansas, Lawrence, Kansas
| | - Maithri Kashipathy
- Protein Structure Laboratory, NIH COBRE in Protein Structure and Function, The University of Kansas, Lawrence, Kansas
| | - Justin Douglas
- Bio-NMR Core Facility, NIH COBRE in Protein Structure and Function, The University of Kansas, Lawrence, Kansas
| | - Philip Gao
- Protein Production Group, NIH COBRE in Protein Structure and Function, The University of Kansas, Lawrence, Kansas
| | - Kevin Battaile
- IMCA-CAT, Hauptman Woodward Medical Research Institute, Argonne, Illinois
| | - Robert Hanzlik
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas
| | - Scott Lovell
- Protein Structure Laboratory, NIH COBRE in Protein Structure and Function, The University of Kansas, Lawrence, Kansas
| | - Liang Xu
- Department of Molecular Biosciences, The University of Kansas, Lawrence, Kansas.,Department of Radiation Oncology, The University of Kansas Cancer Center, Kansas City, Kansas
| |
Collapse
|
18
|
Duggimpudi S, Kloetgen A, Maney SK, Münch PC, Hezaveh K, Shaykhalishahi H, Hoyer W, McHardy AC, Lang PA, Borkhardt A, Hoell JI. Transcriptome-wide analysis uncovers the targets of the RNA-binding protein MSI2 and effects of MSI2's RNA-binding activity on IL-6 signaling. J Biol Chem 2018; 293:15359-15369. [PMID: 30126842 DOI: 10.1074/jbc.ra118.002243] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 07/23/2018] [Indexed: 12/14/2022] Open
Abstract
The RNA-binding protein Musashi 2 (MSI2) has emerged as an important regulator in cancer initiation, progression, and drug resistance. Translocations and deregulation of the MSI2 gene are diagnostic of certain cancers, including chronic myeloid leukemia (CML) with translocation t(7;17), acute myeloid leukemia (AML) with translocation t(10;17), and some cases of B-precursor acute lymphoblastic leukemia (pB-ALL). To better understand the function of MSI2 in leukemia, the mRNA targets that are bound and regulated by MSI2 and their MSI2-binding motifs need to be identified. To this end, using photoactivatable ribonucleoside cross-linking and immunoprecipitation (PAR-CLIP) and the multiple EM for motif elicitation (MEME) analysis tool, here we identified MSI2's mRNA targets and the consensus RNA-recognition element (RRE) motif recognized by MSI2 (UUAG). Of note, MSI2 knockdown altered the expression of several genes with roles in eukaryotic initiation factor 2 (eIF2), hepatocyte growth factor (HGF), and epidermal growth factor (EGF) signaling pathways. We also show that MSI2 regulates classic interleukin-6 (IL-6) signaling by promoting the degradation of the mRNA of IL-6 signal transducer (IL6ST or GP130), which, in turn, affected the phosphorylation statuses of signal transducer and activator of transcription 3 (STAT3) and the mitogen-activated protein kinase ERK. In summary, we have identified multiple MSI2-regulated mRNAs and provided evidence that MSI2 controls IL6ST activity that control oncogenic signaling networks. Our findings may help inform strategies for unraveling the role of MSI2 in leukemia to pave the way for the development of targeted therapies.
Collapse
Affiliation(s)
- Sujitha Duggimpudi
- From the Department of Pediatric Oncology, Hematology and Clinical Immunology, Center for Child and Adolescent Health, Medical Faculty, Heinrich Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Andreas Kloetgen
- From the Department of Pediatric Oncology, Hematology and Clinical Immunology, Center for Child and Adolescent Health, Medical Faculty, Heinrich Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany.,Department of Algorithmic Bioinformatics, Heinrich Heine University, Universitätsstrasse 1, 40225 Düsseldorf, Germany.,Computational Biology of Infection Research, Helmholtz Center for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany, and
| | - Sathish Kumar Maney
- Department of Molecular Medicine II, Heinrich Heine University, Universitätsstrasse 1, 40225, Düsseldorf, Germany
| | - Philipp C Münch
- Department of Algorithmic Bioinformatics, Heinrich Heine University, Universitätsstrasse 1, 40225 Düsseldorf, Germany.,Computational Biology of Infection Research, Helmholtz Center for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany, and
| | - Kebria Hezaveh
- From the Department of Pediatric Oncology, Hematology and Clinical Immunology, Center for Child and Adolescent Health, Medical Faculty, Heinrich Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Hamed Shaykhalishahi
- Institute of Physical Biology, Heinrich Heine University, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Wolfgang Hoyer
- Institute of Physical Biology, Heinrich Heine University, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Alice C McHardy
- Department of Algorithmic Bioinformatics, Heinrich Heine University, Universitätsstrasse 1, 40225 Düsseldorf, Germany.,Computational Biology of Infection Research, Helmholtz Center for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany, and
| | - Philipp A Lang
- Department of Molecular Medicine II, Heinrich Heine University, Universitätsstrasse 1, 40225, Düsseldorf, Germany
| | - Arndt Borkhardt
- From the Department of Pediatric Oncology, Hematology and Clinical Immunology, Center for Child and Adolescent Health, Medical Faculty, Heinrich Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Jessica I Hoell
- From the Department of Pediatric Oncology, Hematology and Clinical Immunology, Center for Child and Adolescent Health, Medical Faculty, Heinrich Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany,
| |
Collapse
|
19
|
Lan L, Liu H, Smith AR, Appelman C, Yu J, Larsen S, Marquez RT, Wu X, Liu FY, Gao P, Gowthaman R, Karanicolas J, De Guzman RN, Rogers S, Aubé J, Neufeld KL, Xu L. Natural product derivative Gossypolone inhibits Musashi family of RNA-binding proteins. BMC Cancer 2018; 18:809. [PMID: 30097032 PMCID: PMC6086024 DOI: 10.1186/s12885-018-4704-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 07/30/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The Musashi (MSI) family of RNA-binding proteins is best known for the role in post-transcriptional regulation of target mRNAs. Elevated MSI1 levels in a variety of human cancer are associated with up-regulation of Notch/Wnt signaling. MSI1 binds to and negatively regulates translation of Numb and APC (adenomatous polyposis coli), negative regulators of Notch and Wnt signaling respectively. METHODS Previously, we have shown that the natural product (-)-gossypol as the first known small molecule inhibitor of MSI1 that down-regulates Notch/Wnt signaling and inhibits tumor xenograft growth in vivo. Using a fluorescence polarization (FP) competition assay, we identified gossypolone (Gn) with a > 20-fold increase in Ki value compared to (-)-gossypol. We validated Gn binding to MSI1 using surface plasmon resonance, nuclear magnetic resonance, and cellular thermal shift assay, and tested the effects of Gn on colon cancer cells and colon cancer DLD-1 xenografts in nude mice. RESULTS In colon cancer cells, Gn reduced Notch/Wnt signaling and induced apoptosis. Compared to (-)-gossypol, the same concentration of Gn is less active in all the cell assays tested. To increase Gn bioavailability, we used PEGylated liposomes in our in vivo studies. Gn-lip via tail vein injection inhibited the growth of human colon cancer DLD-1 xenografts in nude mice, as compared to the untreated control (P < 0.01, n = 10). CONCLUSION Our data suggest that PEGylation improved the bioavailability of Gn as well as achieved tumor-targeted delivery and controlled release of Gn, which enhanced its overall biocompatibility and drug efficacy in vivo. This provides proof of concept for the development of Gn-lip as a molecular therapy for colon cancer with MSI1/MSI2 overexpression.
Collapse
Affiliation(s)
- Lan Lan
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
| | - Hao Liu
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
- Current address: School of Pharmacy, Southwest Medical University, Luzhou City, China
| | - Amber R Smith
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
| | - Carl Appelman
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
| | - Jia Yu
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, China
| | - Sarah Larsen
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
| | - Rebecca T Marquez
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
| | - Xiaoqing Wu
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
| | - Frank Y Liu
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
| | - Philip Gao
- Protein Production Group, NIH COBRE in Protein Structure and Function, Lawrence, USA
| | - Ragul Gowthaman
- Center for Computational Biology, University of Kansas, Lawrence, Kansas, USA
| | - John Karanicolas
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Roberto N De Guzman
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
| | - Steven Rogers
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Jeffrey Aubé
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Kristi L Neufeld
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
| | - Liang Xu
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA.
- Department of Radiation Oncology, University of Kansas Cancer Center, Kansas City, Kansas, USA.
| |
Collapse
|
20
|
Yang S, Sheng L, Xu K, Wang Y, Zhu H, Zhang P, Mu Q, Ouyang G. Anticancer effect of quinacrine on diffuse large B‑cell lymphoma via inhibition of MSI2‑NUMB signaling pathway. Mol Med Rep 2018; 17:522-530. [PMID: 29115587 DOI: 10.3892/mmr.2017.7892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 09/08/2017] [Indexed: 11/05/2022] Open
Abstract
Diffuse large B‑cell lymphoma (DLBCL) is the most common subtype of non‑Hodgkin's lymphoma. Despite improvements in the clinical outcomes of DLBCL, ~30% of patients will develop relapse/refractory disease. Therefore, novel therapeutic drugs have been investigated to improve disease outcomes. Previous studies have revealed the anticancer effects of quinacrine (QC) on tumor cells in vitro, although its role in human DLBCL is yet to be identified. The present study sought to examine the cytotoxic effect of QC on DLBCL cells. QC induced G0/G1 cell cycle arrest and apoptosis in the DLBCL cell lines SU‑DHL‑8 and OCI‑LY01, in a dose‑dependent manner, in addition to the downregulation of cyclin‑dependent kinase 4/6 and the upregulation of cleaved poly‑ADP ribose polymerase 1. Upon exposure to QC, RNA‑binding protein Musashi homolog 2 inactivation and activation of protein numb homolog were observed. In addition, QC was able to inhibit the expression of Myc proto‑oncogene protein. The results of the present study indicated that QC may be a potential anti‑DLBCL drug.
Collapse
Affiliation(s)
- Shujun Yang
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Lixia Sheng
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Kaihong Xu
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Yi Wang
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Huiling Zhu
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Ping Zhang
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Qitian Mu
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Guifang Ouyang
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| |
Collapse
|
21
|
Human oncoprotein Musashi-2 N-terminal RNA recognition motif backbone assignment and identification of RNA-binding pocket. Oncotarget 2017; 8:106587-106597. [PMID: 29290973 PMCID: PMC5739758 DOI: 10.18632/oncotarget.22540] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 10/30/2017] [Indexed: 12/13/2022] Open
Abstract
RNA-binding protein Musashi-2 (MSI2) is a key regulator in stem cells, it is over-expressed in a variety of cancers and its higher expression is associated with poor prognosis. Like Musashi-1, it contains two N-terminal RRMs (RNA-recognition Motifs, also called RBDs (RNA-binding Domains)), RRM1 and RRM2, which mediate the binding to their target mRNAs. Previous studies have obtained the three-dimensional structures of the RBDs of Musashi-1 and the RBD1:RNA complex. Here we show the binding of MSI2-RRM1 to a 15nt Numb RNA in Fluorescence Polarization assay and time resolved Fluorescence Resonance Energy Transfer assay. Using nuclear magnetic resonance (NMR) spectroscopy we assigned the backbone resonances of MSI2-RRM1, and characterized the direct interaction of RRM1 to Numb RNA r(GUAGU). Our NMR titration and structure modeling studies showed that MSI2-RRM1 and MSI1-RBD1 have similar RNA binding events and binding pockets. This work adds significant information to MSI2-RRM1 structure and RNA binding pocket, and contributes to the development of MSI2 specific and MSI1/MSI2 dual inhibitors.
Collapse
|
22
|
Kudinov AE, Karanicolas J, Golemis EA, Boumber Y. Musashi RNA-Binding Proteins as Cancer Drivers and Novel Therapeutic Targets. Clin Cancer Res 2017; 23:2143-2153. [PMID: 28143872 DOI: 10.1158/1078-0432.ccr-16-2728] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 12/16/2016] [Accepted: 12/19/2016] [Indexed: 12/12/2022]
Abstract
Aberrant gene expression that drives human cancer can arise from epigenetic dysregulation. Although much attention has focused on altered activity of transcription factors and chromatin-modulating proteins, proteins that act posttranscriptionally can potently affect expression of oncogenic signaling proteins. The RNA-binding proteins (RBP) Musashi-1 (MSI1) and Musashi-2 (MSI2) are emerging as regulators of multiple critical biological processes relevant to cancer initiation, progression, and drug resistance. Following identification of Musashi as a regulator of progenitor cell identity in Drosophila, the human Musashi proteins were initially linked to control of maintenance of hematopoietic stem cells, then stem cell compartments for additional cell types. More recently, the Musashi proteins were found to be overexpressed and prognostic of outcome in numerous cancer types, including colorectal, lung, and pancreatic cancers; glioblastoma; and several leukemias. MSI1 and MSI2 bind and regulate the mRNA stability and translation of proteins operating in essential oncogenic signaling pathways, including NUMB/Notch, PTEN/mTOR, TGFβ/SMAD3, MYC, cMET, and others. On the basis of these activities, MSI proteins maintain cancer stem cell populations and regulate cancer invasion, metastasis, and development of more aggressive cancer phenotypes, including drug resistance. Although RBPs are viewed as difficult therapeutic targets, initial efforts to develop MSI-specific inhibitors are promising, and RNA interference-based approaches to inhibiting these proteins have had promising outcomes in preclinical studies. In the interim, understanding the function of these translational regulators may yield insight into the relationship between mRNA expression and protein expression in tumors, guiding tumor-profiling analysis. This review provides a current overview of Musashi as a cancer driver and novel therapeutic target. Clin Cancer Res; 23(9); 2143-53. ©2017 AACR.
Collapse
Affiliation(s)
- Alexander E Kudinov
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - John Karanicolas
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Erica A Golemis
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Yanis Boumber
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania. .,Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| |
Collapse
|
23
|
Guo K, Cui J, Quan M, Xie D, Jia Z, Wei D, Wang L, Gao Y, Ma Q, Xie K. The Novel KLF4/MSI2 Signaling Pathway Regulates Growth and Metastasis of Pancreatic Cancer. Clin Cancer Res 2016; 23:687-696. [PMID: 27449499 DOI: 10.1158/1078-0432.ccr-16-1064] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 06/24/2016] [Accepted: 07/13/2016] [Indexed: 02/05/2023]
Abstract
PURPOSE Musashi 2 (MSI2) is reported to be a potential oncoprotein in cases of leukemia and several solid tumors. However, its expression, function, and regulation in pancreatic ductal adenocarcinoma (PDAC) cases have yet to be demonstrated. Therefore, in the current study, we investigated the clinical significance and biologic effects of MSI2 expression in PDAC cases and sought to delineate the clinical significance of the newly identified Krüppel-like factor 4 (KLF4)/MSI2 regulatory pathway. EXPERIMENTAL DESIGN MSI2 expression and its association with multiple clinicopathologic characteristics in human PDAC specimens were analyzed immunohistochemically. The biological functions of MSI2 regarding PDAC cell growth, migration, invasion, and metastasis were studied using gain- and loss-of-function assays both in vitro and in vivo Regulation of MSI2 expression by KLF4 was examined in several cancer cell lines, and the underlying mechanisms were studied using molecular biologic methods. RESULTS MSI2 expression was markedly increased in both PDAC cell lines and human PDAC specimens, and high MSI2 expression was associated with poor prognosis for PDAC. Forced MSI2 expression promoted PDAC proliferation, migration, and invasion in vitro and growth and metastasis in vivo, whereas knockdown of MSI2 expression did the opposite. Transcriptional inhibition of MSI2 expression by KLF4 occurred in multiple PDAC cell lines as well as mouse models of PDAC. CONCLUSIONS Lost expression of KLF4, a transcriptional repressor of MSI2 results in overexpression of MSI2 in PDACs, which may be a biomarker for accurate prognosis. A dysregulated KLF4/MSI2 signaling pathway promotes PDAC progression and metastasis. Clin Cancer Res; 23(3); 687-96. ©2016 AACR.
Collapse
Affiliation(s)
- Kun Guo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China.,Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jiujie Cui
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ming Quan
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Oncology, Shanghai East Hospital, Shanghai Tongji University, Shanghai, P.R. China
| | - Dacheng Xie
- Department of Oncology, Shanghai East Hospital, Shanghai Tongji University, Shanghai, P.R. China
| | - Zhiliang Jia
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daoyan Wei
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Liang Wang
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yong Gao
- Department of Oncology, Shanghai East Hospital, Shanghai Tongji University, Shanghai, P.R. China.
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China.
| | - Keping Xie
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
24
|
Han Y, Ye A, Zhang Y, Cai Z, Wang W, Sun L, Jiang S, Wu J, Yu K, Zhang S. Musashi-2 Silencing Exerts Potent Activity against Acute Myeloid Leukemia and Enhances Chemosensitivity to Daunorubicin. PLoS One 2015; 10:e0136484. [PMID: 26308531 PMCID: PMC4550418 DOI: 10.1371/journal.pone.0136484] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 08/04/2015] [Indexed: 12/22/2022] Open
Abstract
RNA-binding protein Musashi-2 (Msi2) is known to play a critical role in leukemogenesis and contributes to poor clinical prognosis in acute myeloid leukemia (AML). However, the effect of Msi2 silencing on treatment for AML still remains poorly understood. In this study, we used lentivirus-mediated RNA interference targeting Msi2 to investigate the resulting changes in cellular processes and the underlying mechanisms in AML cell lines as well as primary AML cells isolated from AML patients. We found that Msi2 was highly expressed in AML cells, and its depletion inhibited Ki-67 expression and resulted in decreased in vitro and in vivo proliferation. Msi2 silencing induced cell cycle arrest in G0/G1 phase, with decreased Cyclin D1 and increased p21 expression. Msi2 silencing induced apoptosis through down-regulation of Bcl-2 expression and up-regulation of Bax expression. Suppression of Akt, Erk1/2 and p38 phosphorylation also contributed to apoptosis mediated by Msi2 silencing. Finally, Msi2 silencing in AML cells also enhanced their chemosensitivity to daunorubicin. Conclusively, our data suggest that Msi2 is a promising target for gene therapy to optimize conventional chemotherapeutics in AML treatment.
Collapse
MESH Headings
- Antibiotics, Antineoplastic/pharmacology
- Apoptosis/drug effects
- Cell Cycle Checkpoints/drug effects
- Cell Proliferation/drug effects
- Daunorubicin/pharmacology
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Leukemic/drug effects
- Gene Silencing
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- RNA, Small Interfering/genetics
- RNA-Binding Proteins/antagonists & inhibitors
- RNA-Binding Proteins/genetics
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Yixiang Han
- Laboratory of Internal Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, China
| | - Aifang Ye
- Laboratory of Internal Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, China
| | - Yan Zhang
- Key Laboratory of Molecular Medicine, Ministry of Education, and Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, 200032, China
| | - Zhimin Cai
- Laboratory of Internal Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, China
| | - Wei Wang
- Department of Hematology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Lan Sun
- Department of Hematology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Songfu Jiang
- Department of Hematology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Jianbo Wu
- Laboratory of Internal Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, China
| | - Kang Yu
- Department of Hematology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Shenghui Zhang
- Department of Hematology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
- * E-mail:
| |
Collapse
|