1
|
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
|
2
|
Bağcı Ö, Özdemir EM, Şanlıtürk B. Variant Analysis of miRNA Regulatory Genes in 35 Sporadic Lung Carcinoma Tumors. DOKL BIOCHEM BIOPHYS 2023; 513:S1-S7. [PMID: 38472669 DOI: 10.1134/s1607672924600052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 03/14/2024]
Abstract
Lung cancer is one of the cancer types with the highest mortality worldwide. The most frequently mutated genes known to be clinically important in lung cancers are EGFR, BRAF, and KRAS genes. Therefore, the therapeutic agents developed are directed against variants that cause over-activation of the EGFR-KRAS-BRAF-BRAF-MEK/ERK signalling pathway. However, different responses of patients to Tyrosine Kinase Inhibitors (TKIs) suggest that new prognostic biomarkers should be defined and epigenetic mechanisms may be related to this situation. METHODS In this study, sequence analyses of AGO2, DICER, and DROSHA genes involved in miRNA biogenesis and EGFR, KRAS, and BRAF genes were performed in 35 patients with sporadic lung cancer. RESULTS We found variations in genes involved in miRNA biogenesis that have not been previously reported in the literature. In addition, we found 4 different variants in the EGFR gene that have been described in the literature. In addition, a statistically significant association was found between the presence of mutations in at least one of the genes involved in miRNA biogenesis and metastasis (p:0.02). CONCLUSIONS In conclusion, genomic dysregulation of key miRNA biogenesis genes may be one of the possible reasons for the differential response of patients to therapeutic agents and the development of metastasis in EGFR wild type tumours.
Collapse
Affiliation(s)
- Özkan Bağcı
- Department of Medical Genetics, Selcuk University, School of Medicine, Konya, Turkey.
| | | | - Batuhan Şanlıtürk
- Department of Medical Genetics, Selcuk University, School of Medicine, Konya, Turkey
| |
Collapse
|
3
|
Zeng Z, Fu M, Hu Y, Wei Y, Wei X, Luo M. Regulation and signaling pathways in cancer stem cells: implications for targeted therapy for cancer. Mol Cancer 2023; 22:172. [PMID: 37853437 PMCID: PMC10583419 DOI: 10.1186/s12943-023-01877-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 10/05/2023] [Indexed: 10/20/2023] Open
Abstract
Cancer stem cells (CSCs), initially identified in leukemia in 1994, constitute a distinct subset of tumor cells characterized by surface markers such as CD133, CD44, and ALDH. Their behavior is regulated through a complex interplay of networks, including transcriptional, post-transcriptional, epigenetic, tumor microenvironment (TME), and epithelial-mesenchymal transition (EMT) factors. Numerous signaling pathways were found to be involved in the regulatory network of CSCs. The maintenance of CSC characteristics plays a pivotal role in driving CSC-associated tumor metastasis and conferring resistance to therapy. Consequently, CSCs have emerged as promising targets in cancer treatment. To date, researchers have developed several anticancer agents tailored to specifically target CSCs, with some of these treatment strategies currently undergoing preclinical or clinical trials. In this review, we outline the origin and biological characteristics of CSCs, explore the regulatory networks governing CSCs, discuss the signaling pathways implicated in these networks, and investigate the influential factors contributing to therapy resistance in CSCs. Finally, we offer insights into preclinical and clinical agents designed to eliminate CSCs.
Collapse
Affiliation(s)
- Zhen Zeng
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Minyang Fu
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Yuan Hu
- Department of Pediatric Nephrology Nursing, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Min Luo
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China.
| |
Collapse
|
4
|
Moreira ARS, Lim J, Urbaniak A, Banik J, Bronson K, Lagasse A, Hardy L, Haney A, Allensworth M, Miles TK, Gies A, Byrum SD, Wilczynska A, Boehm U, Kharas M, Lengner C, MacNicol MC, Childs GV, MacNicol AM, Odle AK. Musashi Exerts Control of Gonadotrope Target mRNA Translation During the Mouse Estrous Cycle. Endocrinology 2023; 164:bqad113. [PMID: 37477898 PMCID: PMC10402870 DOI: 10.1210/endocr/bqad113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/30/2023] [Accepted: 07/20/2023] [Indexed: 07/22/2023]
Abstract
The anterior pituitary controls key biological processes, including growth, metabolism, reproduction, and stress responses through distinct cell types that each secrete specific hormones. The anterior pituitary cells show a remarkable level of cell type plasticity that mediates the shifts in hormone-producing cell populations that are required to meet organismal needs. The molecular mechanisms underlying pituitary cell plasticity are not well understood. Recent work has implicated the pituitary stem cell populations and specifically, the mRNA binding proteins of the Musashi family in control of pituitary cell type identity. In this study we have identified the target mRNAs that mediate Musashi function in the adult mouse pituitary and demonstrate the requirement for Musashi function in vivo. Using Musashi RNA immunoprecipitation, we identify a cohort of 1184 mRNAs that show specific Musashi binding. Identified Musashi targets include the Gnrhr mRNA, which encodes the gonadotropin-releasing hormone receptor (GnRHR), and the Fshb mRNA, encoding follicle-stimulating hormone (FSH). Reporter assays reveal that Musashi functions to exert repression of translation of the Fshb mRNA, in addition to the previously observed repression of the Gnrhr mRNA. Importantly, mice engineered to lack Musashi in gonadotropes demonstrate a failure to repress translation of the endogenous Gnrhr and Fshb mRNAs during the estrous cycle and display a significant heterogeneity in litter sizes. The range of identified target mRNAs suggests that, in addition to these key gonadotrope proteins, Musashi may exert broad regulatory control over the pituitary proteome in a cell type-specific manner.
Collapse
Affiliation(s)
- Ana Rita Silva Moreira
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Juchan Lim
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Alicja Urbaniak
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jewel Banik
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Katherine Bronson
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Alex Lagasse
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Linda Hardy
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Anessa Haney
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Melody Allensworth
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Tiffany K Miles
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Allen Gies
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Arkansas Children's Research Institute, Arkansas Children's Hospital, Little Rock, AR 72202, USA
| | - Ania Wilczynska
- Bit.bio, The Dorothy Hodgkin Building, Babraham Research Campus, Cambridge CB22 3FH, UK
| | - Ulrich Boehm
- Department of Experimental Pharmacology, Center for Molecular Signaling, Saarland University School of Medicine, Homburg 66421, Germany
| | - Michael Kharas
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Christopher Lengner
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19146, USA
| | - Melanie C MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Gwen V Childs
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Angus M MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Angela K Odle
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
5
|
Knockdown of the stem cell marker Musashi-1 inhibits endometrial cancer growth and sensitizes cells to radiation. Stem Cell Res Ther 2022; 13:212. [PMID: 35619161 PMCID: PMC9137084 DOI: 10.1186/s13287-022-02891-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/02/2022] [Indexed: 11/22/2022] Open
Abstract
Background Endometrial carcinoma is the most common gynecological cancer in Europe. Musashi-1 is known to be a key regulator of endometrial cancer stem cells and a negative prognostic marker. In the present study, we aimed to understand growth and gene expression patterns in endometrial carcinoma after Musashi-1 knockdown in vitro and in vivo. Changes in therapeutic resistance were also assessed.
Methods First, we performed analyses to understand Musashi-1 expression patterns using The Cancer Genome Atlas database. We then proceeded to assess effects of small interfering RNA-based Musashi-1 targeting in two endometrial carcinoma cell lines, Ishikawa and KLE. After quantifying baseline changes in cell metabolism, we used MTT tests to assess chemotherapy effects and colony formation assays to understand changes in radioresistance. For mechanistic study, we used quantitative polymerase chain reaction (qPCR) and western blotting of key Musashi-1 target genes and compared results to primary tissue database studies. Finally, xenograft experiments in a mouse model helped understand in vivo effects of Musashi-1 knockdown. Results Musashi-1 is aberrantly expressed in primary tumor tissues. In vitro, silencing of Musashi-1 resulted in a strong decline in cell proliferation and radioresistance, while chemoresistance remained unchanged. Loss of Musashi-1 led to downregulation of telomerase, DNA-dependent protein kinase, the Notch pathway and overexpression of cyclin-dependent kinase inhibitor p21, the latter of which we identified as a key mediator of Msi-1 knockdown-related anti-proliferative signaling. In vivo, the anti-proliferative effect was confirmed, with Msi-1 knockdown tumors being about 40% reduced in size. Conclusions Musashi-1 knockdown resulted in a strong decrease in endometrial cancer proliferation and a loss of radioresistance, suggesting therapeutic potential. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02891-3.
Collapse
|
6
|
Yang YP, Lee ACL, Lin LT, Chen YW, Huang PI, Ma HI, Chen YC, Lo WL, Lan YT, Fang WL, Wang CY, Liu YY, Hsu PK, Lin WC, Li CP, Chen MT, Chien CS, Wang ML. Strategic Decoy Peptides Interfere with MSI1/AGO2 Interaction to Elicit Tumor Suppression Effects. Cancers (Basel) 2022; 14:cancers14030505. [PMID: 35158774 PMCID: PMC8833744 DOI: 10.3390/cancers14030505] [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: 11/16/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Peptide drugs that can specifically target undesirable protein–protein interactions that lead to oncogenic developments have emerged as the next era of future medicine for cancers. To combat GBM tumor progression, our study offers an alternative therapeutic strategy via targeting the protein–protein interaction between MSI1 and AGO2 with synthetic peptides identified from the C-terminus of MSI1 in peptide arrays. Our present data revealed for the first time that peptidic disruption to the MSI1/AGO2 complex known for promoting cancer stemness and progression could lead to encouraging therapeutic efficacy at both in vitro and in vivo levels. The significantly suppressed tumor growth and prolonged survival rates in PDX tumor models by decoy peptides evidently provided a new rationale for stratifying patients with MSI1/AGO2-targeted therapeutics. Abstract Peptide drugs that target protein–protein interactions have attracted mounting research efforts towards clinical developments over the past decades. Increasing reports have indicated that expression of Musashi 1 (MSI1) is tightly correlated to high grade of cancers as well as enrichment of cancer stem cells. Treatment failure in malignant tumors glioblastoma multiform (GBM) had also been correlated to CSC-regulating properties of MSI1. It is thus imperative to develop new therapeutics that could effectively improve current regimens used in clinics. MSI1 and AGO2 are two emerging oncogenic molecules that both contribute to GBM tumorigenesis through mRNA regulation of targets involved in apoptosis and cell cycle. In this study, we designed peptide arrays covering the C-terminus of MSI1 and identified two peptides (Pep#11 and Pep#26) that could specifically interfere with the binding with AGO2. Our Biacore analyses ascertained binding between the identified peptides and AGO2. Recombinant reporter system Gaussian luciferase and fluorescent bioconjugate techniques were employed to determine biological functions and pharmacokinetic characteristics of these two peptides. Our data suggested that Pep#11 and Pep#26 could function as decoy peptides by mimicking the interaction function of MSI1 with its binding partner AGO2 in vitro and in vivo. Further experiments using GMB animal models corroborated the ability of Pep#11 and Pep#26 in disrupting MSI1/AGO2 interaction and consequently anti-tumorigenicity and prolonged survival rates. These striking therapeutic efficacies orchestrated by the synthetic peptides were attributed to the decoy function to C-terminal MSI1, especially in malignant brain tumors and glioblastoma.
Collapse
Affiliation(s)
- Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan; (Y.-P.Y.); (A.C.-L.L.); (Y.-C.C.)
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-W.C.); (P.-I.H.); (W.-L.L.); (Y.-T.L.); (W.-L.F.); (C.-Y.W.); (Y.-Y.L.); (P.-K.H.); (C.-P.L.); (M.-T.C.)
- Institute of Food Safety and Health Risk Assessment, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Andy Chi-Lung Lee
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan; (Y.-P.Y.); (A.C.-L.L.); (Y.-C.C.)
- Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Liang-Ting Lin
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China;
| | - Yi-Wei Chen
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-W.C.); (P.-I.H.); (W.-L.L.); (Y.-T.L.); (W.-L.F.); (C.-Y.W.); (Y.-Y.L.); (P.-K.H.); (C.-P.L.); (M.-T.C.)
- Department of Neurosurgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Department of Oncology, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Pin-I Huang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-W.C.); (P.-I.H.); (W.-L.L.); (Y.-T.L.); (W.-L.F.); (C.-Y.W.); (Y.-Y.L.); (P.-K.H.); (C.-P.L.); (M.-T.C.)
- Department of Neurosurgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Department of Oncology, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Hsin-I Ma
- Department of Neurological Surgery, Tri-Service General Hospital and National Defense Medical Center, Taipei 114, Taiwan;
| | - Yi-Chen Chen
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan; (Y.-P.Y.); (A.C.-L.L.); (Y.-C.C.)
| | - Wen-Liang Lo
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-W.C.); (P.-I.H.); (W.-L.L.); (Y.-T.L.); (W.-L.F.); (C.-Y.W.); (Y.-Y.L.); (P.-K.H.); (C.-P.L.); (M.-T.C.)
- Division of Oral and Maxillofacial Surgery, Department of Stomatology, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Yuan-Tzu Lan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-W.C.); (P.-I.H.); (W.-L.L.); (Y.-T.L.); (W.-L.F.); (C.-Y.W.); (Y.-Y.L.); (P.-K.H.); (C.-P.L.); (M.-T.C.)
- Division of Colon & Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Wen-Liang Fang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-W.C.); (P.-I.H.); (W.-L.L.); (Y.-T.L.); (W.-L.F.); (C.-Y.W.); (Y.-Y.L.); (P.-K.H.); (C.-P.L.); (M.-T.C.)
- Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Chien-Ying Wang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-W.C.); (P.-I.H.); (W.-L.L.); (Y.-T.L.); (W.-L.F.); (C.-Y.W.); (Y.-Y.L.); (P.-K.H.); (C.-P.L.); (M.-T.C.)
- Division of Trauma, Department of Emergency Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Department of Physical Education and Health, University of Taipei, Taipei 111, Taiwan
| | - Yung-Yang Liu
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-W.C.); (P.-I.H.); (W.-L.L.); (Y.-T.L.); (W.-L.F.); (C.-Y.W.); (Y.-Y.L.); (P.-K.H.); (C.-P.L.); (M.-T.C.)
- Chest Department, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Po-Kuei Hsu
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-W.C.); (P.-I.H.); (W.-L.L.); (Y.-T.L.); (W.-L.F.); (C.-Y.W.); (Y.-Y.L.); (P.-K.H.); (C.-P.L.); (M.-T.C.)
- Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Wen-Chang Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan;
| | - Chung-Pin Li
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-W.C.); (P.-I.H.); (W.-L.L.); (Y.-T.L.); (W.-L.F.); (C.-Y.W.); (Y.-Y.L.); (P.-K.H.); (C.-P.L.); (M.-T.C.)
- Department of Medical Education, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Ming-Teh Chen
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-W.C.); (P.-I.H.); (W.-L.L.); (Y.-T.L.); (W.-L.F.); (C.-Y.W.); (Y.-Y.L.); (P.-K.H.); (C.-P.L.); (M.-T.C.)
- Department of Neurosurgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Department of Medical Education, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Chian-Shiu Chien
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan; (Y.-P.Y.); (A.C.-L.L.); (Y.-C.C.)
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-W.C.); (P.-I.H.); (W.-L.L.); (Y.-T.L.); (W.-L.F.); (C.-Y.W.); (Y.-Y.L.); (P.-K.H.); (C.-P.L.); (M.-T.C.)
- Correspondence: (C.-S.C.); (M.-L.W.); Tel.: +886-2-5568-1156 (M.-L.W.); Fax: +886-2-2875-7435 (M.-L.W.)
| | - Mong-Lien Wang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan; (Y.-P.Y.); (A.C.-L.L.); (Y.-C.C.)
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-W.C.); (P.-I.H.); (W.-L.L.); (Y.-T.L.); (W.-L.F.); (C.-Y.W.); (Y.-Y.L.); (P.-K.H.); (C.-P.L.); (M.-T.C.)
- Institute of Food Safety and Health Risk Assessment, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Correspondence: (C.-S.C.); (M.-L.W.); Tel.: +886-2-5568-1156 (M.-L.W.); Fax: +886-2-2875-7435 (M.-L.W.)
| |
Collapse
|
7
|
Wang S, Wang S, Zhang X, Meng D, Xia Q, Xie S, Shen S, Yu B, Hu J, Liu H, Yan W. Comprehensive analysis of prognosis-related alternative splicing events in ovarian cancer. RNA Biol 2022; 19:1007-1018. [PMID: 35980273 PMCID: PMC9397453 DOI: 10.1080/15476286.2022.2113148] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Ovarian cancer (OV) is characterized by high incidence and poor prognosis. Increasing evidence indicates that aberrant alternative splicing (AS) events are associated with the pathogenesis of cancer. We examined prognosis-related alternative splicing events and constructed a clinically applicable model to predict patients’ outcomes. Public database including The Cancer Genome Atlas (TCGA), TCGA SpliceSeq, and the Genomics of Drug Sensitivity in Cancer databases were used to detect the AS expression, immune cell infiltration and IC50. The prognosis-related AS model was constructed and validated by using Cox regression, LASSO regression, C-index, calibration plots, and ROC curves. A total of eight AS events (including FLT3LG|50942|AP) were selected to establish the prognosis-related AS model. Compared with high-risk group, low-risk group had a better outcome (P = 1.794e-06), was more sensitive to paclitaxel (P = 0.022), and higher proportions of plasma cells. We explored the upstream regulatory mechanisms of prognosis-related AS and found that two splicing factor and 156 tag single nucleotide polymorphisms may be involved in the regulation of prognosis-related AS. In order to assess patient prognosis more comprehensively, we constructed a clinically applicable model combining risk score and clinicopathological features, and the 1 -, and 3-year AUCs of the clinically applicable model were 0.812, and 0.726, which were 7.5% and 3.3% higher than that of the risk score. We constructed a prognostic signature for OV patients and comprehensively analysed the regulatory characteristics of the prognostic AS events in OV.
Collapse
Affiliation(s)
| | - Shiyuan Wang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Xing Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Dan Meng
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Qianqian Xia
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Shuqian Xie
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Siyuan Shen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Bingjia Yu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Jing Hu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Haohan Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Wenjing Yan
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
8
|
Liu X, Chen JY, Chien Y, Yang YP, Chen MT, Lin LT. Overview of the molecular mechanisms of migration and invasion in glioblastoma multiforme. J Chin Med Assoc 2021; 84:669-677. [PMID: 34029218 DOI: 10.1097/jcma.0000000000000552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma (GBM) is one of the most devastating cancers, with an approximate median survival of only 16 months. Although some new insights into the fantastic heterogeneity of this kind of brain tumor have been revealed in recent studies, all subclasses of GBM still demonstrate highly aggressive invasion properties to the surrounding parenchyma. This behavior has become the main obstruction to current curative therapies as invasive GBM cells migrate away from these foci after surgical therapies. Therefore, this review aimed to provide a relatively comprehensive study of GBM invasion mechanisms, which contains an intricate network of interactions and signaling pathways with the extracellular matrix (ECM). Among these related molecules, TGF-β, the ECM, Akt, and microRNAs are most significant in terms of cellular procedures related to GBM motility and invasion. Moreover, we also review data indicating that Musashi-1 (MSI1), a neural RNA-binding protein (RBP), regulates GBM motility and invasion, maintains stem cell populations in GBM, and promotes drug-resistant GBM phenotypes by stimulating necessary oncogenic signaling pathways through binding and regulating mRNA stability. Importantly, these necessary oncogenic signaling pathways have a close connection with TGF-β, ECM, and Akt. Thus, it appears promising to find MSI-specific inhibitors or RNA interference-based treatments to prevent the actions of these molecules despite using RBPs, which are known as hard therapeutic targets. In summary, this review aims to provide a better understanding of these signaling pathways to help in developing novel therapeutic approaches with better outcomes in preclinical studies.
Collapse
Affiliation(s)
- Xian Liu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Ju-Yu Chen
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Yueh Chien
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Ming-Teh Chen
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Medical Education & Department of Neurosurgery, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Liang-Ting Lin
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
- Department of Health Technology and Informatics, Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, Guangdong, China
| |
Collapse
|
9
|
Bley N, Hmedat A, Müller S, Rolnik R, Rausch A, Lederer M, Hüttelmaier S. Musashi-1-A Stemness RBP for Cancer Therapy? BIOLOGY 2021; 10:407. [PMID: 34062997 PMCID: PMC8148009 DOI: 10.3390/biology10050407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 04/29/2021] [Accepted: 05/01/2021] [Indexed: 12/12/2022]
Abstract
The RNA-binding protein Musashi-1 (MSI1) promotes stemness during development and cancer. By controlling target mRNA turnover and translation, MSI1 is implicated in the regulation of cancer hallmarks such as cell cycle or Notch signaling. Thereby, the protein enhanced cancer growth and therapy resistance to standard regimes. Due to its specific expression pattern and diverse functions, MSI1 represents an interesting target for cancer therapy in the future. In this review we summarize previous findings on MSI1's implications in developmental processes of other organisms. We revisit MSI1's expression in a set of solid cancers, describe mechanistic details and implications in MSI1 associated cancer hallmark pathways and highlight current research in drug development identifying the first MSI1-directed inhibitors with anti-tumor activity.
Collapse
Affiliation(s)
- Nadine Bley
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
- Core Facility Imaging, Institute for Molecular Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany
| | - Ali Hmedat
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
| | - Simon Müller
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
| | - Robin Rolnik
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
| | - Alexander Rausch
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
- Core Facility Imaging, Institute for Molecular Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany
| | - Marcell Lederer
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
| | - Stefan Hüttelmaier
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
| |
Collapse
|
10
|
Forouzanfar M, Lachinani L, Dormiani K, Nasr-Esfahani MH, Ghaedi K. Increased expression of MUSASHI1 in epithelial breast cancer cells is due to down regulation of miR-125b. BMC Mol Cell Biol 2021; 22:10. [PMID: 33541259 PMCID: PMC7863248 DOI: 10.1186/s12860-021-00348-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 01/26/2021] [Indexed: 01/04/2023] Open
Abstract
Background Musashi1 (MSI1) is an oncogenic protein with a crucial role in the proliferation and characteristics of the epithelial cells in breast cancer. The change in expression of MSI1 has a role in solid tumor progression. There are different factors that regulate MSI1 expression in various cancer tissues including microRNAs which are considered as one of the most important of these factors. The aim of our study is identification of the molecular cause of maximal expression of MSI1 in epithelial breast cancer cell lines. Results Among predicted microRNAs, miR-125b, miR-637 and miR-802 were able to significantly reduce the luciferase activity. In addition, the relative expression of these three miRNAs were measured in the cancerous cell lines that results showed a significant reduction in expression of all microRNAs. On the other hand, only the overexpression of miR-125b caused a change in the expression pattern of MSI1 in breast epithelial cancer cell lines. Accordingly, our results demonstrated that the exogenous expression of miR-125b decreased not only the MSI1 protein but also expression of epithelial markers in breast cancer cells. Conclusions The results of luciferase reporter assay showed that MSI1 is a direct target for miR-125b in epithelial breast cancer cells. Moreover, higher amount of MSI1 in those cell lines seems due to the reduced amount of miR-125b, which is responsible for epithelial features of those kinds of cancer cells. Therefore, the modulation of miR-125b may be a potential approach to help to combat against epithelial breast tumors. Supplementary Information The online version contains supplementary material available at 10.1186/s12860-021-00348-8.
Collapse
Affiliation(s)
- Mahboobeh Forouzanfar
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar Jerib Ave., Azadi Square, Isfahan, P.O. Code 81746, Iran
| | - Liana Lachinani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, P.O. Code 816513-1378, Iran
| | - Kianoush Dormiani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, P.O. Code 816513-1378, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, P.O. Code 816513-1378, Iran.
| | - Kamran Ghaedi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar Jerib Ave., Azadi Square, Isfahan, P.O. Code 81746, Iran.
| |
Collapse
|
11
|
MSI1 Promotes the Expression of the GBM Stem Cell Marker CD44 by Impairing miRNA-Dependent Degradation. Cancers (Basel) 2020; 12:cancers12123654. [PMID: 33291443 PMCID: PMC7762192 DOI: 10.3390/cancers12123654] [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: 11/12/2020] [Revised: 11/30/2020] [Accepted: 12/03/2020] [Indexed: 01/03/2023] Open
Abstract
Simple Summary Glioblastoma (GBM) is the most lethal brain tumor with a median survival rate of approximately 14 months. GBM patients commonly suffer from tumor recurrence, indicating that populations of chemo/radio-resistant stem cell-like tumor cells survive treatments. Here we reveal that the neuronal stem cell marker Musashi1 (MSI1) is highly expressed in primary GBM and recurrences. We identify a novel regulatory role of MSI1 in GBM-derived cell lines and patient-derived tumorspheres, the enhancement of stemness marker expression, here demonstrated for CD44. Furthermore, we provide a rationale for MSI1-centered therapeutic targeting strategies to improve treatment options of this chemo/radio-resistant malignancy. Abstract The stem cell marker Musashi1 (MSI1) is highly expressed during neurogenesis and in glioblastoma (GBM). MSI1 promotes self-renewal and impairs differentiation in cancer and non-malignant progenitor cells. However, a comprehensive understanding of its role in promoting GBM-driving networks remains to be deciphered. We demonstrate that MSI1 is highly expressed in GBM recurrences, an oncologist’s major defiance. For the first time, we provide evidence that MSI1 promotes the expression of stem cell markers like CD44, co-expressed with MSI1 within recurrence-promoting cells at the migrating front of primary GBM samples. With GBM cell models of pediatric and adult origin, including isolated primary tumorspheres, we show that MSI1 promotes stem cell-like characteristics. Importantly, it impairs CD44 downregulation in a 3′UTR- and miRNA-dependent manner by controlling mRNA turnover. This regulation is disturbed by the previously reported MSI1 inhibitor luteolin, providing further evidence for a therapeutic target potential of MSI1 in GBM treatment.
Collapse
|
12
|
Forouzanfar M, Lachinani L, Dormiani K, Nasr-Esfahani MH, Gure AO, Ghaedi K. Intracellular functions of RNA-binding protein, Musashi1, in stem and cancer cells. Stem Cell Res Ther 2020; 11:193. [PMID: 32448364 PMCID: PMC7245930 DOI: 10.1186/s13287-020-01703-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/31/2020] [Accepted: 05/05/2020] [Indexed: 12/13/2022] Open
Abstract
RNA-binding protein, musashi1 (MSI1), is a main protein in asymmetric cell division of the sensory organ precursor cells, whereas its expression is reported to be upregulated in cancers. This protein is a critical element in proliferation of stem and cancer stem cells, which acts through Wnt and Notch signaling pathways. Moreover, MSI1 modulates malignancy and chemoresistance of lung cancer cells via activating the Akt signaling. Due to the main role of MSI1 in metastasis and cancer development, MSI1 would be an appropriate candidate for cancer therapy. Downregulation of MSI1 inhibits proliferation of cancer stem cells and reduces the growth of solid tumors in several cancers. On the other hand, MSI1 expression is regulated by microRNAs in such a way that several different tumor suppressor miRNAs negatively regulate oncogenic MSI1 and inhibit migration and tumor metastasis. The aim of this review is summarizing the role of MSI1 in stem cell proliferation and cancer promotion.
Collapse
Affiliation(s)
- Mahboobeh Forouzanfar
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar Jerib Ave., Azadi Square, Isfahan, P.O. Code 81746, Iran
| | - Liana Lachinani
- Department of Molecular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, P.O. Code 816513-1378, Iran
| | - Kianoush Dormiani
- Department of Molecular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, P.O. Code 816513-1378, Iran.
| | - Mohammad Hossein Nasr-Esfahani
- Department of Molecular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, P.O. Code 816513-1378, Iran. .,Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| | - Ali Osmay Gure
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey
| | - Kamran Ghaedi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar Jerib Ave., Azadi Square, Isfahan, P.O. Code 81746, Iran. .,Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| |
Collapse
|