1
|
Mao Z, Wang B, Chen Y, Ying J, Wang H, Li J, Zhang C, Zhuo J. Musashi orchestrates melanism in Laodelphax striatellus. INSECT SCIENCE 2024. [PMID: 38706046 DOI: 10.1111/1744-7917.13372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/23/2024] [Accepted: 03/25/2024] [Indexed: 05/07/2024]
Abstract
In insects, melanism, a fundamental pigmentation process, is of significant importance in evolutionary biology due to its complex genetic foundation. We investigated the role of the RNA-binding gene Musashi (msi) in melanism in Laodelphax striatellus, a Hemiptera species. We identified a single L. striatellus msi homolog, Lsmsi, encoding a 357 amino acid protein with 2 RNA recognition motifs. RNA interference-mediated knockdown of LsMsi resulted in complete body melanism and increased cuticular permeability. Additionally, we found the involvement of G protein-coupled receptor A42 and tyrosine hydroxylase (Th) in L. striatellus melanism. Knockdown of LsTh lightened the epidermis, showing dehydration signs, while LsA42 knockdown enhanced LsTh expression, leading to melanism. Surprisingly, Lsmsi knockdown decreased both LsA42 and LsTh expression, which was expected to cause whitening but resulted in melanism. Further, we found that Lsmsi influenced downstream genes like phenoloxidase homolog LsPo and dopa decarboxylase (Ddc) homolog LsDdc in the tyrosine-mediated melanism pathway. Extending to Nilaparvata lugens and Sogatella furcifera, we demonstrated the conserved role of msi in melanism among Delphacidae. Given MSI proteins' roles in cancer and tumors in vertebrates, our study is the first to link msi in insects to Delphacidae body color melanization via the tyrosine-mediated pathway, offering fresh perspectives on the genetic basis of insect melanism and msi functions.
Collapse
Affiliation(s)
- Zeping Mao
- State Key Laboratory for ManagingBiotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Institute of Plant Virology, Ningbo University, Ningbo, Zhejiang Province, 315211, China
| | - Biyun Wang
- State Key Laboratory for ManagingBiotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Institute of Plant Virology, Ningbo University, Ningbo, Zhejiang Province, 315211, China
| | - Youyuan Chen
- State Key Laboratory for ManagingBiotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Institute of Plant Virology, Ningbo University, Ningbo, Zhejiang Province, 315211, China
| | - Jinjun Ying
- State Key Laboratory for ManagingBiotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Institute of Plant Virology, Ningbo University, Ningbo, Zhejiang Province, 315211, China
| | - Haiqiang Wang
- State Key Laboratory for ManagingBiotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Institute of Plant Virology, Ningbo University, Ningbo, Zhejiang Province, 315211, China
| | - Junmin Li
- State Key Laboratory for ManagingBiotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Institute of Plant Virology, Ningbo University, Ningbo, Zhejiang Province, 315211, China
| | - Chuanxi Zhang
- State Key Laboratory for ManagingBiotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Institute of Plant Virology, Ningbo University, Ningbo, Zhejiang Province, 315211, China
| | - Jichong Zhuo
- State Key Laboratory for ManagingBiotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Institute of Plant Virology, Ningbo University, Ningbo, Zhejiang Province, 315211, China
| |
Collapse
|
2
|
Lv Y, Wen L, Hu WJ, Deng C, Ren HW, Bao YN, Su BW, Gao P, Man ZY, Luo YY, Li CJ, Xiang ZX, Wang B, Luan ZL. Schizophrenia in the genetic era: a review from development history, clinical features and genomic research approaches to insights of susceptibility genes. Metab Brain Dis 2024; 39:147-171. [PMID: 37542622 DOI: 10.1007/s11011-023-01271-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/27/2023] [Indexed: 08/07/2023]
Abstract
Schizophrenia is a devastating neuropsychiatric disorder affecting 1% of the world population and ranks as one of the disorders providing the most severe burden for society. Schizophrenia etiology remains obscure involving multi-risk factors, such as genetic, environmental, nutritional, and developmental factors. Complex interactions of genetic and environmental factors have been implicated in the etiology of schizophrenia. This review provides an overview of the historical origins, pathophysiological mechanisms, diagnosis, clinical symptoms and corresponding treatment of schizophrenia. In addition, as schizophrenia is a polygenic, genetic disorder caused by the combined action of multiple micro-effective genes, we further detail several approaches, such as candidate gene association study (CGAS) and genome-wide association study (GWAS), which are commonly used in schizophrenia genomics studies. A number of GWASs about schizophrenia have been performed with the hope to identify novel, consistent and influential risk genetic factors. Finally, some schizophrenia susceptibility genes have been identified and reported in recent years and their biological functions are also listed. This review may serve as a summary of past research on schizophrenia genomics and susceptibility genes (NRG1, DISC1, RELN, BDNF, MSI2), which may point the way to future schizophrenia genetics research. In addition, depending on the above discovery of susceptibility genes and their exact function, the development and application of antipsychotic drugs will be promoted in the future.
Collapse
Affiliation(s)
- Ye Lv
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Lin Wen
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Wen-Juan Hu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Chong Deng
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027, China
| | - Hui-Wen Ren
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Ya-Nan Bao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Bo-Wei Su
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Ping Gao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Zi-Yue Man
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Yi-Yang Luo
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Cheng-Jie Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Zhi-Xin Xiang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Bing Wang
- Department of Endocrinology and Metabolism, The Central hospital of Dalian University of Technology, Dalian, 116000, China.
| | - Zhi-Lin Luan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
3
|
Yu Q, Zhang Y, Tian Y, Peng A, Cui X, Ding B, Yang L, Liu Y, Ju Y, Gao C. Exosomal Circ_FMN2 Derived from the Serum of Colorectal Cancer Patients Promotes Cancer Progression by miR-338-3p/MSI1 Axis. Appl Biochem Biotechnol 2023; 195:7322-7337. [PMID: 36995659 DOI: 10.1007/s12010-023-04456-3] [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] [Accepted: 03/15/2023] [Indexed: 03/31/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is a common malignancy of the gastrointestinal tract with high incidence and mortality. Exosomal circular RNA (circRNA) has been shown to be associated with the malignant progression of cancers, including CRC. Circ_0005100 (named as circ_FMN2) has been shown to promote CRC cell proliferation and migration. However, whether exosomal circ_FMN2 participated in CRC progression remains unclear. METHODS Exosomes were isolated from the serum of CRC patients and then identified using transmission electron microscope. Western blot assay was used to test the protein levels of exosome markers, proliferation-related marker, metastasis-related markers and musashi-1 (MSI1). The expression levels of circ_FMN2, microRNA (miR)-338-3p and MSI1 were detected by qPCR. Flow cytometry, colony formation assay, MTT assay, and transwell assay were employed to measure cell cycle, apoptosis, colony formation ability, viability, migration and invasion. Dual-luciferase reporter assay was performed to assess the interaction between miR-338-3p and circ_FMN2 or MSI1. BALB/c nude mice was used to conduct animal experiments. RESULTS Circ_FMN2 was overexpressed in the exosomes of CRC patient's serums and CRC cells. Overexpressed exosomal circ_FMN2 could promote CRC cell proliferation, metastasis, and suppress apoptosis. Circ_FMN2 acted as miR-338-3p sponge. MiR-338-3p overexpression reversed the promotion effect of circ_FMN2 on CRC progression. MSI1 was found to be a target of miR-338-3p, and its overexpression revoked the inhibitory effect of miR-338-3p on CRC progression. Furthermore, exosomal circ_FMN2 overexpression also could facilitate CRC tumor growth in vivo. CONCLUSION Exosomal circ_FMN2 accelerated CRC progression through miR-338-3p/MSI1 axis, revealing that exosomal circ_FMN2 might be a target for CRC treatment.
Collapse
Affiliation(s)
- Qiyao Yu
- Department of Research, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yi Zhang
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050011, Hebei Province, China
| | - Yanming Tian
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050011, Hebei Province, China
| | - Ale Peng
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, No. 12, Jian Kang Road, Shijiazhuang, 050011, Hebei Province, China
| | - Xiujing Cui
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, No. 12, Jian Kang Road, Shijiazhuang, 050011, Hebei Province, China
| | - Boyue Ding
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, No. 12, Jian Kang Road, Shijiazhuang, 050011, Hebei Province, China
| | - Lei Yang
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, No. 12, Jian Kang Road, Shijiazhuang, 050011, Hebei Province, China
| | - Yabin Liu
- Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yingchao Ju
- Department of experimental animal center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chao Gao
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, No. 12, Jian Kang Road, Shijiazhuang, 050011, Hebei Province, China.
| |
Collapse
|
4
|
Zhang T, Wu S, Xu R, Zhang S, Wang M, Li J. Musashi-2 binds with Fbxo6 to induce Rnaset2 ubiquitination and chemokine signaling pathway during vascular smooth muscle cell phenotypic switch in atherosclerosis. Cell Signal 2023; 111:110869. [PMID: 37633478 DOI: 10.1016/j.cellsig.2023.110869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/11/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
OBJECTIVE The objective of this study is to determine how Musashi-2 (MSI2) affects vascular smooth muscle cell (VSMC) phenotypic switch and contributes to atherosclerosis (AS). METHODS Primary mouse VSMCs were transfected with MSI2 specific siRNA and treated with platelet-derived growth factor-BB (PDGF-BB). The proliferation, cell-cycle, and migration of VSMCs were determined by CCK-8, flow cytometry, wound healing, and transwell assays. Western blot and qRT-PCR were conducted to analyze the protein and mRNA expression. Moreover, the correlation between MSI2, Fbxo6, Rnaset2, and chemokine signaling was predicted and verified using RNAct database, KEGG, wiki, RNA-binding protein immunoprecipitation and co-immunoprecipitation. Moreover, H&E and Oil Red O staining were employed for assessing necrotic core and lipid accumulation in AS mouse aorta tissues. The numbers of B lymphocytes and monocytes, and the levels of triglyceride (TG), total cholesterol (TC), high-density lipoprotein cholesterol (HDLC), and low-density lipoprotein cholesterol (LDL-C) in AS mice blood were investigated using flow cytometry and corresponding commercial kits, respectively. RESULTS MSI2 was up-regulated in the PDGF-BB-treated VSMCs. Knockdown of MSI2 inhibited VSMC proliferation, cell-cycle, and migration. Moreover, MSI2 regulated VSMC phenotypic switch through binding with Fbxo6 to induce Rnaset2 ubiquitination. MSI2 knockdown inhibited chemokine signaling via regulating Fbxo6/Rnaset2 axis. In AS mice, knockdown of MSI2 inhibited the formation of necrotic core and atherosclerotic plaque, and inhibited chemokine signaling via regulating Fbxo6/Rnaset2 axis. CONCLUSION Our findings demonstrated that MSI2 could bind with Fbxo6 to induce Rnaset2 ubiquitination and the activation of chemokine signaling pathway during VSMC phenotypic switch in AS.
Collapse
Affiliation(s)
- Tao Zhang
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, Shandong, China; Department of General Surgery, The First Hospital Affiliated with Shandong First Medical University, Jinan 250014, Shandong, China
| | - Shusheng Wu
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, Shandong, China; Department of General Surgery, The First Hospital Affiliated with Shandong First Medical University, Jinan 250014, Shandong, China
| | - Rongwei Xu
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, Shandong, China; Department of General Surgery, The First Hospital Affiliated with Shandong First Medical University, Jinan 250014, Shandong, China
| | - Shuguang Zhang
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, Shandong, China; Department of General Surgery, The First Hospital Affiliated with Shandong First Medical University, Jinan 250014, Shandong, China
| | - Minghai Wang
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, Shandong, China; Department of General Surgery, The First Hospital Affiliated with Shandong First Medical University, Jinan 250014, Shandong, China.
| | - Jie Li
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, Shandong, China; Department of General Surgery, The First Hospital Affiliated with Shandong First Medical University, Jinan 250014, Shandong, China.
| |
Collapse
|
5
|
Chagas PS, Veronez LC, de Sousa GR, Cruzeiro GAV, Corrêa CAP, Saggioro FP, de Paula Queiroz RG, Marie SKN, Brandalise SR, Cardinalli IA, Yunes JA, Júnior CGC, Machado HR, Santos MV, Scrideli CA, Tone LG, Valera ET. Musashi-1 regulates cell cycle and confers resistance to cisplatin treatment in Group 3/4 medulloblastomas cells. Hum Cell 2023; 36:2129-2139. [PMID: 37460706 DOI: 10.1007/s13577-023-00954-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/10/2023] [Indexed: 10/20/2023]
Abstract
Groups (Grp) 3 and 4 are aggressive molecular subgroups of medulloblastoma (MB), with high rates of leptomeningeal dissemination. To date, there is still a paucity of biomarkers for these subtypes of MBs. In this study, we investigated the clinical significance and biological functions of Musashi-1 (MSI1) in Grp3 and Grp4-MBs. First, we assessed the expression profile of MSI1 in 59 primary MB samples (15-WNT, 18-SHH, 9-Grp3, and 17-Grp4 subgroups) by qRT-PCR. MSI1 mRNA expression levels were also validated in an additional public dataset of MBs (GSE85217). The ROC curve was used to validate the diagnostic standards of MSI1 expression. Next, the potential correlated cell-cycle genes were measured by RNA-Seq. Cell cycle, cell viability, and apoptosis were evaluated in a Grp3/Grp4 MB cell line after knockdown of MSI1 and cisplatin treatment. We identified an overexpression of MSI1 with a high accuracy to discriminate Grp3/Grp4-MBs from non-Grp3/Grp4-MBs. We identified that MSI1 knockdown not only triggered transcriptional changes in the cell-cycle pathway, but also affected G2/M phase in vitro, supporting the role of knockdown of MSI1 in cell-cycle arrest. Finally, MSI1 knockdown decreased cell viability and sensitized D283-Med cells to cisplatin treatment by enhancing cell apoptosis. Based on these findings, we suggest that MSI1 modulates cell-cycle progression and may play a role as biomarker for Grp3/Grp4-MBs. In addition, MSI1 knockdown combined with cisplatin may offer a potential strategy to be further explored in Grp3/Grp4-MBs.
Collapse
Affiliation(s)
- Pablo Shimaoka Chagas
- Department of Genetics, Ribeirão Preto Medical School-University of São Paulo, Bandeirantes Avenue, 3900, Ribeirão Preto, São Paulo, 14048-900, Brazil.
| | - Luciana Chain Veronez
- Department of Pediatrics, Clinics Hospital-Ribeirão Preto Medical School-University of São Paulo, Ribeirão Preto, Brazil
| | - Graziella Ribeiro de Sousa
- Department of Genetics, Ribeirão Preto Medical School-University of São Paulo, Bandeirantes Avenue, 3900, Ribeirão Preto, São Paulo, 14048-900, Brazil
| | - Gustavo Alencastro Veiga Cruzeiro
- Department of Pediatrics, Clinics Hospital-Ribeirão Preto Medical School-University of São Paulo, Ribeirão Preto, Brazil
- Department of Pediatric Oncology, Harvard Medical School-Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Carolina Alves Pereira Corrêa
- Department of Pediatrics, Clinics Hospital-Ribeirão Preto Medical School-University of São Paulo, Ribeirão Preto, Brazil
| | - Fabiano Pinto Saggioro
- Department of Pathology, Ribeirão Preto Medical School, 3900 Bandeirantes Avenue, Ribeirão Preto, SP, 14049-900, Brazil
- Department of Pathology, Rede D'Or São Luiz Hospital, Rua das Perobas, São Paulo, SP, 04321-120, Brazil
| | - Rosane Gomes de Paula Queiroz
- Department of Pediatrics, Clinics Hospital-Ribeirão Preto Medical School-University of São Paulo, Ribeirão Preto, Brazil
| | - Suely Kazue Nagahashi Marie
- Laboratory of Cellular and Molecular Biology, Department of Neurology, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | | | | | | - Hélio Rubens Machado
- Division of Pediatric Neurosurgery, Department of Surgery and Anatomy, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Marcelo Volpon Santos
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Bandeirantes Av., Ribeirão Preto, SP, 390014049-900, Brazil
| | - Carlos Alberto Scrideli
- Department of Genetics, Ribeirão Preto Medical School-University of São Paulo, Bandeirantes Avenue, 3900, Ribeirão Preto, São Paulo, 14048-900, Brazil
- Department of Pediatrics, Clinics Hospital-Ribeirão Preto Medical School-University of São Paulo, Ribeirão Preto, Brazil
| | - Luiz Gonzaga Tone
- Department of Genetics, Ribeirão Preto Medical School-University of São Paulo, Bandeirantes Avenue, 3900, Ribeirão Preto, São Paulo, 14048-900, Brazil
- Department of Pediatrics, Clinics Hospital-Ribeirão Preto Medical School-University of São Paulo, Ribeirão Preto, Brazil
| | - Elvis Terci Valera
- Department of Pediatrics, Clinics Hospital-Ribeirão Preto Medical School-University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
6
|
Brisset M, Mehlen P, Meurette O, Hollande F. Notch receptor/ligand diversity: contribution to colorectal cancer stem cell heterogeneity. Front Cell Dev Biol 2023; 11:1231416. [PMID: 37860822 PMCID: PMC10582728 DOI: 10.3389/fcell.2023.1231416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/21/2023] [Indexed: 10/21/2023] Open
Abstract
Cancer cell heterogeneity is a key contributor to therapeutic failure and post-treatment recurrence. Targeting cell subpopulations responsible for chemoresistance and recurrence seems to be an attractive approach to improve treatment outcome in cancer patients. However, this remains challenging due to the complexity and incomplete characterization of tumor cell subpopulations. The heterogeneity of cells exhibiting stemness-related features, such as self-renewal and chemoresistance, fuels this complexity. Notch signaling is a known regulator of cancer stem cell (CSC) features in colorectal cancer (CRC), though the effects of its heterogenous signaling on CRC cell stemness are only just emerging. In this review, we discuss how Notch ligand-receptor specificity contributes to regulating stemness, self-renewal, chemoresistance and cancer stem cells heterogeneity in CRC.
Collapse
Affiliation(s)
- Morgan Brisset
- Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, The University of Melbourne, Melbourne, VIC, Australia
- Cancer Cell Death Laboratory, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Patrick Mehlen
- Cancer Cell Death Laboratory, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Olivier Meurette
- Cancer Cell Death Laboratory, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Frédéric Hollande
- Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
7
|
Haiduk TS, Sicking M, Brücksken KA, Espinoza-Sánchez NA, Eder KM, Kemper B, Eich HT, Götte M, Greve B, Troschel FM. Dysregulated Stem Cell Markers Musashi-1 and Musashi-2 are Associated with Therapy Resistance in Inflammatory Breast Cancer. Arch Med Res 2023; 54:102855. [PMID: 37481823 DOI: 10.1016/j.arcmed.2023.102855] [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: 01/20/2023] [Revised: 06/21/2023] [Accepted: 07/11/2023] [Indexed: 07/25/2023]
Abstract
BACKGROUND AND AIM While preliminary evidence points to pro-tumorigenic roles for the Musashi (MSI) RNA-binding proteins Musashi-1 (MSI1) and Musashi-2 (MSI2) in some breast cancer subtypes, no data exist for inflammatory breast cancer (IBC). METHODS MSI gene expression was quantified in IBC SUM149PT cells. We then used small interfering RNA-based MSI1 and MSI2 double knockdown (DKD) to understand gene expression and functional changes upon MSI depletion. We characterized cancer stem cell characteristics, cell apoptosis and cell cycle progression via flow cytometry, mammospheres via spheroid assays, migration and proliferation via digital holographic microscopy, and cell viability using BrdU assays. Chemoresistance was determined for paclitaxel and cisplatin with MTT assays and radioresistance was assessed with clonogenic analyses. In parallel, we supported our in vitro data by analyzing publicly available patient IBC gene expression datasets. RESULTS MSI1 and MSI2 are upregulated in breast cancer generally and IBC specifically. MSI2 is more commonly expressed compared to MSI1. MSI DKD attenuated proliferation, cell cycle progression, migration, and cell viability while increasing apoptosis. Stem cell characteristics CD44(+)/CD24(-), TERT and Oct4 were associated with MSI expression in vivo and were decreased in vitro after MSI DKD as was ALDH expression and mammosphere formation. In vivo, chemoresistant tumors were characterized by MSI upregulation upon chemotherapy application. In vitro, MSI DKD was able to alleviate chemo- and radioresistance. CONCLUSIONS The Musashi RNA binding proteins are dysregulated in IBC and associated with tumor proliferation, cancer stem cell phenotype, chemo- and radioresistance. MSI downregulation alleviates therapy resistance and attenuates tumor proliferation in vitro.
Collapse
Affiliation(s)
- Tiffany S Haiduk
- Department of Radiation Oncology, University Hospital Münster, Münster, Germany
| | - Mark Sicking
- Department of Radiation Oncology, University Hospital Münster, Münster, Germany
| | - Kathrin A Brücksken
- Department of Radiation Oncology, University Hospital Münster, Münster, Germany
| | - Nancy A Espinoza-Sánchez
- Department of Radiation Oncology, University Hospital Münster, Münster, Germany; Department of Gynecology and Obstetrics, University Hospital Münster, Münster, Germany
| | - Kai Moritz Eder
- Biomedical Technology Center, Medical Faculty, University of Münster, Münster, Germany
| | - Björn Kemper
- Biomedical Technology Center, Medical Faculty, University of Münster, Münster, Germany
| | - Hans Theodor Eich
- Department of Radiation Oncology, University Hospital Münster, Münster, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, University Hospital Münster, Münster, Germany
| | - Burkhard Greve
- Department of Radiation Oncology, University Hospital Münster, Münster, Germany
| | - Fabian M Troschel
- Department of Radiation Oncology, University Hospital Münster, Münster, Germany.
| |
Collapse
|
8
|
Unzueta-Larrinaga P, Barrena-Barbadillo R, Ibarra-Lecue I, Horrillo I, Villate A, Recio M, Meana JJ, Diez-Alarcia R, Mentxaka O, Segarra R, Etxebarria N, Callado LF, Urigüen L. Isolation and Differentiation of Neurons and Glial Cells from Olfactory Epithelium in Living Subjects. Mol Neurobiol 2023; 60:4472-4487. [PMID: 37118325 PMCID: PMC10293402 DOI: 10.1007/s12035-023-03363-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 04/19/2023] [Indexed: 04/30/2023]
Abstract
The study of psychiatric and neurological diseases requires the substrate in which the disorders occur, that is, the nervous tissue. Currently, several types of human bio-specimens are being used for research, including postmortem brains, cerebrospinal fluid, induced pluripotent stem (iPS) cells, and induced neuronal (iN) cells. However, these samples are far from providing a useful predictive, diagnostic, or prognostic biomarker. The olfactory epithelium is a region close to the brain that has received increased interest as a research tool for the study of brain mechanisms in complex neuropsychiatric and neurological diseases. The olfactory sensory neurons are replaced by neurogenesis throughout adult life from stem cells on the basement membrane. These stem cells are multipotent and can be propagated in neurospheres, proliferated in vitro and differentiated into multiple cell types including neurons and glia. For all these reasons, olfactory epithelium provides a unique resource for investigating neuronal molecular markers of neuropsychiatric and neurological diseases. Here, we describe the isolation and culture of human differentiated neurons and glial cells from olfactory epithelium of living subjects by an easy and non-invasive exfoliation method that may serve as a useful tool for the research in brain diseases.
Collapse
Affiliation(s)
- Paula Unzueta-Larrinaga
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Rocío Barrena-Barbadillo
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Department of Nursery, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Inés Ibarra-Lecue
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Igor Horrillo
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain
| | - Aitor Villate
- Department of Analytical Chemistry, University of the Basque Country UPV/EHU, Leioa, Spain
- PiE-UPV/EHU, Plentzia, ItsasEstazioa, Areatza Pasealekua, 48620, Plentzia, Spain
| | - Maria Recio
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Department of Psychiatry, Cruces University Hospital, Barakaldo, Spain
| | - J Javier Meana
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain
| | - Rebeca Diez-Alarcia
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain
| | - Oihane Mentxaka
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Department of Psychiatry, Cruces University Hospital, Barakaldo, Spain
- Department of Neurosciences, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Rafael Segarra
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain
- Department of Psychiatry, Cruces University Hospital, Barakaldo, Spain
- Department of Neurosciences, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Nestor Etxebarria
- Department of Analytical Chemistry, University of the Basque Country UPV/EHU, Leioa, Spain
- PiE-UPV/EHU, Plentzia, ItsasEstazioa, Areatza Pasealekua, 48620, Plentzia, Spain
| | - Luis F Callado
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain
| | - Leyre Urigüen
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain.
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain.
| |
Collapse
|
9
|
(-)- Gossypol Inhibition of Musashi-Mediated Forgetting Improves Memory and Age-Dependent Memory Decline in Caenorhabditis elegans. Mol Neurobiol 2023; 60:820-835. [PMID: 36378468 PMCID: PMC9849318 DOI: 10.1007/s12035-022-03116-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/03/2022] [Indexed: 11/17/2022]
Abstract
Musashi RNA-binding proteins (MSIs) retain a pivotal role in stem cell maintenance, tumorigenesis, and nervous system development. Recently, we showed in C. elegans that Musashi (MSI-1) actively promotes forgetting upon associative learning via a 3'UTR-dependent translational expression of the Arp2/3 actin branching complex. Here, we investigated the evolutionary conserved role of MSI proteins and the effect of their pharmacological inhibition on memory. Expression of human Musashi 1 (MSI1) and Musashi 2 (MSI2) under the endogenous Musashi promoter fully rescued the phenotype of msi-1(lf) worms. Furthermore, pharmacological inhibition of human MSI1 and MSI2 activity using (-)- gossypol resulted in improved memory retention, without causing locomotor, chemotactic, or learning deficits. No drug effect was observed in msi-1(lf) treated worms. Using Western blotting and confocal microscopy, we found no changes in MSI-1 protein abundance following (-)- gossypol treatment, suggesting that Musashi gene expression remains unaltered and that the compound exerts its inhibitory effect post-translationally. Additionally, (-)- gossypol suppressed the previously seen rescue of the msi-1(lf) phenotype in worms expressing human MSI1 specifically in the AVA neuron, indicating that (-)- gossypol can regulate the Musashi pathway in a memory-related neuronal circuit in worms. Finally, treating aged worms with (-)- gossypol reversed physiological age-dependent memory decline. Taken together, our findings indicate that pharmacological inhibition of Musashi might represent a promising approach for memory modulation.
Collapse
|
10
|
Emerging Roles of RNA-Binding Proteins in Inner Ear Hair Cell Development and Regeneration. Int J Mol Sci 2022; 23:ijms232012393. [PMID: 36293251 PMCID: PMC9604452 DOI: 10.3390/ijms232012393] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/07/2022] [Accepted: 10/14/2022] [Indexed: 11/05/2022] Open
Abstract
RNA-binding proteins (RBPs) regulate gene expression at the post-transcriptional level. They play major roles in the tissue- and stage-specific expression of protein isoforms as well as in the maintenance of protein homeostasis. The inner ear is a bi-functional organ, with the cochlea and the vestibular system required for hearing and for maintaining balance, respectively. It is relatively well documented that transcription factors and signaling pathways are critically involved in the formation of inner ear structures and in the development of hair cells. Accumulating evidence highlights emerging functions of RBPs in the post-transcriptional regulation of inner ear development and hair cell function. Importantly, mutations of splicing factors of the RBP family and defective alternative splicing, which result in inappropriate expression of protein isoforms, lead to deafness in both animal models and humans. Because RBPs are critical regulators of cell proliferation and differentiation, they present the potential to promote hair cell regeneration following noise- or ototoxin-induced damage through mitotic and non-mitotic mechanisms. Therefore, deciphering RBP-regulated events during inner ear development and hair cell regeneration can help define therapeutic strategies for treatment of hearing loss. In this review, we outline our evolving understanding of the implications of RBPs in hair cell formation and hearing disease with the aim of promoting future research in this field.
Collapse
|
11
|
Mastrandreas P, Boglari C, Arnold A, Peter F, de Quervain DJF, Papassotiropoulos A, Stetak A. Phosphorylation of MSI-1 is implicated in the regulation of associative memory in Caenorhabditis elegans. PLoS Genet 2022; 18:e1010420. [PMID: 36223338 PMCID: PMC9555661 DOI: 10.1371/journal.pgen.1010420] [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: 05/02/2022] [Accepted: 09/09/2022] [Indexed: 11/07/2022] Open
Abstract
The Musashi family of RNA-binding proteins controls several biological processes including stem cell maintenance, cell division and neural function. Previously, we demonstrated that the C. elegans Musashi ortholog, msi-1, regulates forgetting via translational repression of the Arp2/3 actin-branching complex. However, the mechanisms controlling MSI-1 activity during the regulation of forgetting are currently unknown. Here we investigated the effects of protein phosphorylation on MSI-1 activity. We showed that MSI-1 function is likely controlled by alterations of its activity rather than its expression levels. Furthermore, we found that MSI-1 is phosphorylated and using mass spectrometry we identified MSI-1 phosphorylation at three residues (T18, S19 and S34). CRISPR-based manipulations of MSI-1 phosphorylation sites revealed that phosphorylation is necessary for MSI-1 function in both short- and long-term aversive olfactory associative memory. Thus, our study provides insight into the mechanisms regulating memory-related MSI-1 activity and may facilitate the development of novel therapeutic approaches. Understanding neural circuits and molecular mechanisms underlying learning and memory are the major challenges of neuroscience. It is a generally accepted model that a learning event causes modification of synapses; strengthening some within a circuit and weakening others (termed “synaptic plasticity”). A plastic nervous system requires not only the ability to acquire and store but also to forget new inputs. While learning and memory is widely investigated, clear-cut evidence for mechanisms involved in forgetting is still sparse. Previously, we demonstrated the role of the protein Musashi (MSI-1) in the active regulation of forgetting in the nematode C. elegans. Here we investigated the role of protein modification (phosphorylation) as a possible regulatory mechanism of the MSI-1 protein activity. We found that MSI-1 protein is modified at different positions and all of these modifications at the protein level contribute to the correct activity of the protein leading to active forgetting of short and long-term memories.
Collapse
Affiliation(s)
- Pavlina Mastrandreas
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Molecular Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- Biozentrum, Life Sciences Training Facility, University of Basel, Basel, Switzerland
| | - Csaba Boglari
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Molecular Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- Biozentrum, Life Sciences Training Facility, University of Basel, Basel, Switzerland
| | - Andreas Arnold
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Molecular Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- Biozentrum, Life Sciences Training Facility, University of Basel, Basel, Switzerland
| | - Fabian Peter
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Molecular Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- Biozentrum, Life Sciences Training Facility, University of Basel, Basel, Switzerland
| | - Dominique J.-F. de Quervain
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Cognitive Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- University Psychiatric Clinics, University of Basel, Basel, Switzerland
| | - Andreas Papassotiropoulos
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Molecular Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- Biozentrum, Life Sciences Training Facility, University of Basel, Basel, Switzerland
- University Psychiatric Clinics, University of Basel, Basel, Switzerland
| | - Attila Stetak
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Molecular Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- Biozentrum, Life Sciences Training Facility, University of Basel, Basel, Switzerland
- University Psychiatric Clinics, University of Basel, Basel, Switzerland
- * E-mail:
| |
Collapse
|
12
|
Kozole J, Heydn R, Wirkert E, Küspert S, Aigner L, Bruun TH, Bogdahn U, Peters S, Johannesen S. Direct Potential Modulation of Neurogenic Differentiation Markers by Granulocyte-Colony Stimulating Factor (G-CSF) in the Rodent Brain. Pharmaceutics 2022; 14:pharmaceutics14091858. [PMID: 36145606 PMCID: PMC9504319 DOI: 10.3390/pharmaceutics14091858] [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: 07/26/2022] [Revised: 08/22/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
The hematopoietic granulocyte-colony stimulating growth factor (G-CSF, filgrastim) is an approved drug in hematology and oncology. Filgrastim's potential in neurodegenerative disorders is gaining increasingly more attention, as preclinical and early clinical studies suggest it could be a promising treatment option. G-CSF has had a tremendous record as a safe drug for more than three decades; however, its effects upon the central nervous system (CNS) are still not fully understood. In contrast to conceptual long-term clinical application with lower dosing, our present pilot study intends to give a first insight into the molecular effects of a single subcutaneous (s.c.) high-dose G-CSF application upon different regions of the rodent brain. We analyzed mRNA-and in some instances-protein data of neurogenic and non-neurogenic differentiation markers in different regions of rat brains five days after G-CSF (1.3 mg/kg) or physiological saline. We found a continuous downregulation of several markers in most brain regions. Remarkably, cerebellum and hypothalamus showed an upregulation of different markers. In conclusion, our study reveals minor suppressive or stimulatory effects of a single exceptional high G-CSF dose upon neurogenic and non-neurogenic differentiation markers in relevant brain regions, excluding unregulated responses or unexpected patterns of marker expression.
Collapse
Affiliation(s)
- Judith Kozole
- Department of Neurology, University Hospital Regensburg, 93053 Regensburg, Germany
- Department of Anesthesiology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Rosmarie Heydn
- Department of Neurology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Eva Wirkert
- Department of Neurology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Sabrina Küspert
- Department of Neurology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, 5020 Salzburg, Austria
| | - Tim-Henrik Bruun
- Department of Neurology, University Hospital Regensburg, 93053 Regensburg, Germany
- Velvio GmbH, 93053 Regensburg, Germany
| | - Ulrich Bogdahn
- Department of Neurology, University Hospital Regensburg, 93053 Regensburg, Germany
- Velvio GmbH, 93053 Regensburg, Germany
- Correspondence: or
| | - Sebastian Peters
- Department of Neurology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Siw Johannesen
- Department of Neurology, University Hospital Regensburg, 93053 Regensburg, Germany
- Department of Neurology, BG Trauma Center, 82418 Murnau (Staffelsee), Germany
| |
Collapse
|
13
|
The Role of RNA-Binding Proteins in Hematological Malignancies. Int J Mol Sci 2022; 23:ijms23179552. [PMID: 36076951 PMCID: PMC9455611 DOI: 10.3390/ijms23179552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/18/2022] [Accepted: 08/21/2022] [Indexed: 11/17/2022] Open
Abstract
Hematological malignancies comprise a plethora of different neoplasms, such as leukemia, lymphoma, and myeloma, plus a myriad of dysplasia, such as myelodysplastic syndromes or anemias. Despite all the advances in patient care and the development of new therapies, some of these malignancies remain incurable, mainly due to resistance and refractoriness to treatment. Therefore, there is an unmet clinical need to identify new biomarkers and potential therapeutic targets that play a role in treatment resistance and contribute to the poor outcomes of these tumors. RNA-binding proteins (RBPs) are a diverse class of proteins that interact with transcripts and noncoding RNAs and are involved in every step of the post-transcriptional processing of transcripts. Dysregulation of RBPs has been associated with the development of hematological malignancies, making them potential valuable biomarkers and potential therapeutic targets. Although a number of dysregulated RBPs have been identified in hematological malignancies, there is a critical need to understand the biology underlying their contribution to pathology, such as the spatiotemporal context and molecular mechanisms involved. In this review, we emphasize the importance of deciphering the regulatory mechanisms of RBPs to pinpoint novel therapeutic targets that could drive or contribute to hematological malignancy biology.
Collapse
|
14
|
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
|
15
|
Suo J, Zou S, Wang J, Han Y, Zhang L, Lv C, Jiang B, Ren Q, Chen L, Yang L, Ji P, Zheng X, Hu P, Zou W. The RNA-binding protein Musashi2 governs osteoblast-adipocyte lineage commitment by suppressing PPARγ signaling. Bone Res 2022; 10:31. [PMID: 35301280 PMCID: PMC8930990 DOI: 10.1038/s41413-022-00202-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/29/2021] [Accepted: 01/08/2022] [Indexed: 11/12/2022] Open
Abstract
Osteoporosis caused by aging is characterized by reduced bone mass and accumulated adipocytes in the bone marrow cavity. How the balance between osteoblastogenesis and adipogenesis from bone marrow mesenchymal stem cells (BMSCs) is lost upon aging is still unclear. Here, we found that the RNA-binding protein Musashi2 (Msi2) regulates BMSC lineage commitment. Msi2 is commonly enriched in stem cells and tumor cells. We found that its expression was downregulated during adipogenic differentiation and upregulated during osteogenic differentiation of BMSCs. Msi2 knockout mice exhibited decreased bone mass with substantial accumulation of marrow adipocytes, similar to aging-induced osteoporosis. Depletion of Msi2 in BMSCs led to increased adipocyte commitment. Transcriptional profiling analysis revealed that Msi2 deficiency led to increased PPARγ signaling. RNA-interacting protein immunoprecipitation assays demonstrated that Msi2 could inhibit the translation of the key adipogenic factor Cebpα, thereby inhibiting PPAR signaling. Furthermore, the expression of Msi2 decreased significantly during the aging process of mice, indicating that decreased Msi2 function during aging contributes to abnormal accumulation of adipocytes in bone marrow and osteoporosis. Thus, our results provide a putative biochemical mechanism for aging-related osteoporosis, suggesting that modulating Msi2 function may benefit the treatment of bone aging.
Collapse
Affiliation(s)
- Jinlong Suo
- Department of Orthopedic Surgery and Institute of Microsurgery on Extremities, Shanghai Jiaotong University Affiliated Sixth People's Hospital, 200233, Shanghai, China
| | - Sihai Zou
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, 401147, Chongqing, China
| | - Jinghui Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yujiao Han
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Lingli Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Chenchen Lv
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Bo Jiang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Qian Ren
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Long Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Lele Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Ping Ji
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, 401147, Chongqing, China
| | - Xianyou Zheng
- Department of Orthopedic Surgery and Institute of Microsurgery on Extremities, Shanghai Jiaotong University Affiliated Sixth People's Hospital, 200233, Shanghai, China.
| | - Ping Hu
- Guangzhou Laboratory, No. 9 XingDaoHuan Road, Guanghzou International Bio lsland, 510005, Guangzhou, China. .,Colorectal Cancer Center/Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China. .,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China. .,Bio-Research Innovation Center, Shanghai Institute of Biochemistry and Cell Biology, Suzhou, China.
| | - Weiguo Zou
- Department of Orthopedic Surgery and Institute of Microsurgery on Extremities, Shanghai Jiaotong University Affiliated Sixth People's Hospital, 200233, Shanghai, China. .,State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China.
| |
Collapse
|
16
|
Fan B, Zhang Q, Wang N, Wang G. LncRNAs, the Molecules Involved in Communications With Colorectal Cancer Stem Cells. Front Oncol 2022; 12:811374. [PMID: 35155247 PMCID: PMC8829571 DOI: 10.3389/fonc.2022.811374] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/07/2022] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer stem cells (CRCSCs) can actively self-renew, as well as having multidirectional differentiation and tumor regeneration abilities. Because the high functional activities of CRCSCs are associated with low cure rates in patients with colorectal cancer, efforts have sought to determine the function and regulatory mechanisms of CRCSCs. To date, however, the potential regulatory mechanisms of CRCSCs remain incompletely understood. Many non-coding genes are involved in tumor invasion and spread through their regulation of CRCSCs, with long non-coding RNAs (lncRNAs) being important non-coding RNAs. LncRNAs may be involved in the colorectal cancer development and drug resistance through their regulation of CRCSCs. This review systematically evaluates the latest research on the ability of lncRNAs to regulate CRCSC signaling pathways and the involvement of these lncRNAs in colorectal cancer promotion and suppression. The regulatory network of lncRNAs in the CRCSC signaling pathway has been determined. Further analysis of the potential clinical applications of lncRNAs as novel clinical diagnostic and prognostic biomarkers and therapeutic targets for colorectal cancer may provide new ideas and protocols for the prevention and treatment of colorectal cancer.
Collapse
Affiliation(s)
- Boyang Fan
- Department of Colorectal Cancer Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qian Zhang
- Department of Colorectal Surgery, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Ning Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Guiyu Wang
- Department of Colorectal Cancer Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
17
|
Zhang W, Liu L, Zhao S, Chen L, Wei Y, Chen W, Ge F. Research progress on RNA‑binding proteins in breast cancer (Review). Oncol Lett 2022; 23:121. [PMID: 35261635 PMCID: PMC8867207 DOI: 10.3892/ol.2022.13241] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 02/03/2022] [Indexed: 11/28/2022] Open
Abstract
Breast cancer is the most common malignancy among women, and the abnormal regulation of gene expression serves an important role in its occurrence and development. However, the molecular mechanisms underlying gene expression are highly complex and heterogeneous, and RNA-binding proteins (RBPs) are among the key regulatory factors. RBPs bind targets in an environment-dependent or environment-independent manner to influence mRNA stability and the translation of genes involved in the formation, progression, metastasis and treatment of breast cancer. Due to the growing interest in these regulators, the present review summarizes the most influential studies concerning RBPs associated with breast cancer to elucidate the role of RBPs in breast cancer and to assess how they interact with other key pathways to provide new molecular targets for the diagnosis and treatment of breast cancer.
Collapse
Affiliation(s)
- Wenzhu Zhang
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Linlin Liu
- School of Forensic Medicine, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Shengdi Zhao
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Liang Chen
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Yuxian Wei
- Department of Endocrine Breast Surgery, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Wenlin Chen
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650118, P.R. China
| | - Fei Ge
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
18
|
Liu F, Yang H, Zhang X, Sun X, Zhou J, Li Y, Liu Y, Zhuang Z, Wang G. Inhibition of Musashi-1 enhances chemotherapeutic sensitivity in gastric cancer patient-derived xenografts. Exp Biol Med (Maywood) 2022; 247:868-879. [PMID: 35135374 DOI: 10.1177/15353702221076793] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Musashi-1 (MSI1), a neural RNA-binding protein, is considered a gastric and intestinal stem cell marker. Although the function of MSI1 in gastric cancer has attracted increasing interest, it is not known whether MSI1 can be used as a biomarker to monitor gastric cancer development and response to treatment. Here, the role of MSI1 in the chemotherapeutic sensitivity of gastric cancer was investigated. Patients with high MSI1 levels had poor outcomes, implicating the gene in the development and progression of the disease. We overexpressed and silenced MSI1 in the human gastric cancer cell lines MKN45 and HGC27, finding that knockdown reduced proliferation, invasion, and migration, while promoting apoptosis. A patient-derived xenograft gastric cancer model was constructed in which mice received chemical drugs, si-MSI1, or a drug-si-MSI1 combination. It was found that blocking MSI1 expression reduced gastric cancer drug tolerance. The combination treatment with si-MSI1 was superior to 5F-dUMP and cisplatin, either separately or in combination, indicating that including si-MSI1 was better than drug therapy alone. Transcriptome sequencing analysis showed that MSI1 altered cell cycle regulation and growth signal transduction, including that of blood vessel epicardial substance (BVES). These results suggest that MSI1 reduces the tolerance of gastric cancer to chemical drugs through modulation of MSI1/BVES signaling.
Collapse
Affiliation(s)
- Fan Liu
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China.,Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China.,Department of Oncology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Huan Yang
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Xinyu Zhang
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Xianglin Sun
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Jiamin Zhou
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Yuan Li
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Yifei Liu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Zhixiang Zhuang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Guohua Wang
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| |
Collapse
|
19
|
The RNA-Binding Protein Musashi1 Regulates a Network of Cell Cycle Genes in Group 4 Medulloblastoma. Cells 2021; 11:cells11010056. [PMID: 35011618 PMCID: PMC8750343 DOI: 10.3390/cells11010056] [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: 09/21/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 11/17/2022] Open
Abstract
Medulloblastoma is the most common malignant brain tumor in children. Treatment with surgery, irradiation, and chemotherapy has improved survival in recent years, but patients are frequently left with devastating neurocognitive and other sequelae. Patients in molecular subgroups 3 and 4 still experience a high mortality rate. To identify new pathways contributing to medulloblastoma development and create new routes for therapy, we have been studying oncogenic RNA-binding proteins. We defined Musashi1 (Msi1) as one of the main drivers of medulloblastoma development. The high expression of Msi1 is prevalent in Group 4 and correlates with poor prognosis while its knockdown disrupted cancer-relevant phenotypes. Genomic analyses (RNA-seq and RIP-seq) indicated that cell cycle and division are the main biological categories regulated by Msi1 in Group 4 medulloblastoma. The most prominent Msi1 targets include CDK2, CDK6, CCND1, CDKN2A, and CCNA1. The inhibition of Msi1 with luteolin affected the growth of CHLA-01 and CHLA-01R Group 4 medulloblastoma cells and a synergistic effect was observed when luteolin and the mitosis inhibitor, vincristine, were combined. These findings indicate that a combined therapeutic strategy (Msi1 + cell cycle/division inhibitors) could work as an alternative to treat Group 4 medulloblastoma.
Collapse
|
20
|
Kawai M, Imaizumi K, Ishikawa M, Shibata S, Shinozaki M, Shibata T, Hashimoto S, Kitagawa T, Ago K, Kajikawa K, Shibata R, Kamata Y, Ushiba J, Koga K, Furue H, Matsumoto M, Nakamura M, Nagoshi N, Okano H. Long-term selective stimulation of transplanted neural stem/progenitor cells for spinal cord injury improves locomotor function. Cell Rep 2021; 37:110019. [PMID: 34818559 DOI: 10.1016/j.celrep.2021.110019] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/06/2021] [Accepted: 10/28/2021] [Indexed: 02/07/2023] Open
Abstract
In cell transplantation therapy for spinal cord injury (SCI), grafted human induced pluripotent stem cell-derived neural stem/progenitor cells (hiPSC-NS/PCs) mainly differentiate into neurons, forming synapses in a process similar to neurodevelopment. In the developing nervous system, the activity of immature neurons has an important role in constructing and maintaining new synapses. Thus, we investigate how enhancing the activity of transplanted hiPSC-NS/PCs affects both the transplanted cells themselves and the host tissue. We find that chemogenetic stimulation of hiPSC-derived neural cells enhances cell activity and neuron-to-neuron interactions in vitro. In a rodent model of SCI, consecutive and selective chemogenetic stimulation of transplanted hiPSC-NS/PCs also enhances the expression of synapse-related genes and proteins in surrounding host tissues and prevents atrophy of the injured spinal cord, thereby improving locomotor function. These findings provide a strategy for enhancing activity within the graft to improve the efficacy of cell transplantation therapy for SCI.
Collapse
Affiliation(s)
- Momotaro Kawai
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Kent Imaizumi
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Mitsuru Ishikawa
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Shinsuke Shibata
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Munehisa Shinozaki
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Takahiro Shibata
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Shogo Hashimoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Takahiro Kitagawa
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Kentaro Ago
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Keita Kajikawa
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Reo Shibata
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Yasuhiro Kamata
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Junichi Ushiba
- Department of Biosciences and Informatics, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku, Yokohama 223-8522, Japan
| | - Keisuke Koga
- Department of Neurophysiology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| | - Hidemasa Furue
- Department of Neurophysiology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| | - Morio Matsumoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Narihito Nagoshi
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan.
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan.
| |
Collapse
|
21
|
Dual Knockdown of Musashi RNA-Binding Proteins MSI-1 and MSI-2 Attenuates Putative Cancer Stem Cell Characteristics and Therapy Resistance in Ovarian Cancer Cells. Int J Mol Sci 2021; 22:ijms222111502. [PMID: 34768932 PMCID: PMC8584030 DOI: 10.3390/ijms222111502] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/14/2021] [Accepted: 10/21/2021] [Indexed: 01/06/2023] Open
Abstract
In ovarian cancer, therapy resistance mechanisms complicate cancer cell eradication. Targeting Musashi RNA-binding proteins (MSI) may increase therapeutic efficacy. Database analyses were performed to identify gene expression associations between MSI proteins and key therapy resistance and cancer stem cell (CSC) genes. Then, ovarian cancer cells were subjected to siRNA-based dual knockdown of MSI-1 and MSI-2. CSC and cell cycle gene expression was investigated using quantitative polymerase chain reaction (qPCR), western blots, and flow cytometry. Metabolic activity and chemoresistance were assessed by MTT assay. Clonogenic assays were used to quantify cell survival post-irradiation. Database analyses demonstrated positive associations between MSI proteins and putative CSC markers NOTCH, MYC, and ALDH4A1 and negative associations with NOTCH inhibitor NUMB. MSI-2 expression was negatively associated with the apoptosis regulator p21. MSI-1 and MSI-2 were positively correlated, informing subsequent dual knockdown experiments. After MSI silencing, CSC genes were downregulated, while cell cycle progression was reduced. Metabolic activity was decreased in some cancer cells. Both chemo- and radioresistance were reduced after dual knockdown, suggesting therapeutic potential. Dual knockdown of MSI proteins is a promising venue to impede tumor growth and sensitize ovarian cancer cells to irradiation and chemotherapy.
Collapse
|
22
|
Abstract
Somatic stem cells are distinguished by their capacity to regenerate themselves and also to produce daughter cells that will differentiate. Self-renewal is achieved through the process of asymmetric cell division which helps to sustain tissue morphogenesis as well as maintain homeostasis. Asymmetric cell division results in the development of two daughter cells with different fates after a single mitosis. Only one daughter cell maintains "stemness" while the other differentiates and achieves a non-stem cell fate. Stem cells also have the capacity to undergo symmetric division of cells that results in the development of two daughter cells which are identical. Symmetric division results in the expansion of the stem cell population. Imbalances and deregulations in these processes can result in diseases such as cancer. Adult mammary stem cells (MaSCs) are a group of cells that play a critical role in the expansion of the mammary gland during puberty and any subsequent pregnancies. Furthermore, given the relatively long lifespans and their capability to undergo self-renewal, adult stem cells have been suggested as ideal candidates for transformation events that lead to the development of cancer. With the possibility that MaSCs can act as the source cells for distinct breast cancer types; understanding their regulation is an important field of research. In this review, we discuss asymmetric cell division in breast/mammary stem cells and implications on further research. We focus on the background history of asymmetric cell division, asymmetric cell division monitoring techniques, identified molecular mechanisms of asymmetric stem cell division, and the role asymmetric cell division may play in breast cancer.
Collapse
Affiliation(s)
| | - Brian W Booth
- Department of Bioengineering, Head-Cellular Engineering Laboratory, 401-1 Rhodes Engineering Research Center, Clemson University, Clemson, SC, 29634, USA.
| |
Collapse
|
23
|
Kamte YS, Chandwani MN, Michaels AC, O’Donnell LA. Neural Stem Cells: What Happens When They Go Viral? Viruses 2021; 13:v13081468. [PMID: 34452333 PMCID: PMC8402908 DOI: 10.3390/v13081468] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/22/2021] [Accepted: 06/28/2021] [Indexed: 12/25/2022] Open
Abstract
Viruses that infect the central nervous system (CNS) are associated with developmental abnormalities as well as neuropsychiatric and degenerative conditions. Many of these viruses such as Zika virus (ZIKV), cytomegalovirus (CMV), and herpes simplex virus (HSV) demonstrate tropism for neural stem cells (NSCs). NSCs are the multipotent progenitor cells of the brain that have the ability to form neurons, astrocytes, and oligodendrocytes. Viral infections often alter the function of NSCs, with profound impacts on the growth and repair of the brain. There are a wide spectrum of effects on NSCs, which differ by the type of virus, the model system, the cell types studied, and the age of the host. Thus, it is a challenge to predict and define the consequences of interactions between viruses and NSCs. The purpose of this review is to dissect the mechanisms by which viruses can affect survival, proliferation, and differentiation of NSCs. This review also sheds light on the contribution of key antiviral cytokines in the impairment of NSC activity during a viral infection, revealing a complex interplay between NSCs, viruses, and the immune system.
Collapse
|
24
|
Talebi-Yazdabadi Z, Jahanbakhsh N, Dormiani K, Forouzanfar M, Lachinani L, Zohrabi D, Tavalaee M, Nasr-Esfahani MH. Assessment of MUSASHI 1 and MUSASHI 2 expression in spermatozoa and testicular tissue. Andrologia 2021; 53:e14187. [PMID: 34309875 DOI: 10.1111/and.14187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/25/2021] [Accepted: 06/24/2021] [Indexed: 01/13/2023] Open
Abstract
MUSASHI (MSI) family plays the main role in the spermatogenesis process. The purpose of this study was the assessment of sperm MSI1 and MSI2, and sperm functional tests in infertile men (n = 30) with varicocele and fertile men (n = 30). Furthermore, MSI1 and MSI2 proteins were assessed in testicular tissue of azoospermic men (n = 9) as well as epididymal spermatozoa and testis of mice. Expression of MSI1 and MSI2 was assessed at RNA and protein levels in human spermatozoa. Sperm concentration and motility were significantly lower, while abnormal sperm morphology, lipid peroxidation, DNA fragmentation and protamine deficiency were significantly higher in men with varicocele compared to fertile individuals. Any significant difference was not observed in the expression of MSI1 and MSI2 mRNA between the two groups. Unlike MSI1 protein that was not detectable in humans, the relative expression of MSI2 protein was similar in varicocele and fertile individuals. The expression level of both Msi1 and Msi2 proteins was also observable in mouse spermatozoa. No significant relationship was observed between sperm functional parameters with expression of these genes. The data of this study demonstrated that although MSI1 and MSI2 play important roles during spermatogenesis, their relative expression in spermatozoa was not affected by varicocele.
Collapse
Affiliation(s)
- Zohreh Talebi-Yazdabadi
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.,ACECR Institute of Higher Education, Isfahan, Iran
| | - Neda Jahanbakhsh
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.,Department of Biology, Faculty of Science, NourDanesh Institute of Higher Education, Isfahan, Iran
| | - Kianoush Dormiani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mahboobeh Forouzanfar
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Liana Lachinani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Dina Zohrabi
- ACECR Institute of Higher Education, Isfahan, Iran
| | - Marziyeh Tavalaee
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.,Isfahan Fertility and Infertility Center, Isfahan, Iran
| |
Collapse
|
25
|
Troschel FM, Palenta H, Borrmann K, Heshe K, Hua SH, Yip GW, Kiesel L, Eich HT, Götte M, Greve B. Knockdown of the prognostic cancer stem cell marker Musashi-1 decreases radio-resistance while enhancing apoptosis in hormone receptor-positive breast cancer cells via p21 WAF1/CIP1. J Cancer Res Clin Oncol 2021; 147:3299-3312. [PMID: 34291358 PMCID: PMC8484224 DOI: 10.1007/s00432-021-03743-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/13/2021] [Indexed: 02/02/2023]
Abstract
Purpose While the stem cell marker Musashi-1 (MSI-1) has been identified as a key player in a wide array of malignancies, few findings exist on its prognostic relevance and relevance for cancer cell death and therapy resistance in breast cancer. Methods First, we determined prognostic relevance of MSI-1 in database analyses regarding multiple survival outcomes. To substantiate findings, MSI-1 was artificially downregulated in MCF-7 breast cancer cells and implications for cancer stem cell markers, cell apoptosis and apoptosis regulator p21, proliferation and radiation response were analyzed via flow cytometry and colony formation. Radiation-induced p21 expression changes were investigated using a dataset containing patient samples obtained before and after irradiation and own in vitro experiments. Results MSI-1 is a negative prognostic marker for disease-free and distant metastasis-free survival in breast cancer and tends to negatively influence overall survival. MSI-1 knockdown downregulated stem cell gene expression and proliferation, but increased p21 levels and apoptosis. Similar to the MSI-1 knockdown effect, p21 expression was strongly increased after irradiation and was expressed at even higher levels in MSI-1 knockdown cells after irradiation. Finally, combined use of MSI-1 silencing and irradiation reduced cancer cell survival. Conclusion MSI-1 is a prognostic marker in breast cancer. MSI-1 silencing downregulates proliferation while increasing apoptosis. The anti-proliferation mediator p21 was upregulated independently after both MSI-1 knockdown and irradiation and even more after both treatments combined, suggesting synergistic potential. Radio-sensitization effects after combining radiation and MSI-1 knockdown underline the potential of MSI-1 as a therapeutic target. Supplementary Information The online version contains supplementary material available at 10.1007/s00432-021-03743-y.
Collapse
Affiliation(s)
- Fabian M Troschel
- Department of Radiation Oncology, University Hospital Münster, 48149, Münster, Germany.
| | - Heike Palenta
- Department of Gynecology and Obstetrics, University Hospital Münster, 48149, Münster, Germany
| | - Katrin Borrmann
- Department of Radiation Oncology, University Hospital Münster, 48149, Münster, Germany
| | - Kristin Heshe
- Department of Radiation Oncology, University Hospital Münster, 48149, Münster, Germany
| | - San Hue Hua
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117594, Singapore
| | - George W Yip
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117594, Singapore
| | - Ludwig Kiesel
- Department of Gynecology and Obstetrics, University Hospital Münster, 48149, Münster, Germany
| | - Hans Theodor Eich
- Department of Radiation Oncology, University Hospital Münster, 48149, Münster, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, University Hospital Münster, 48149, Münster, Germany
| | - Burkhard Greve
- Department of Radiation Oncology, University Hospital Münster, 48149, Münster, Germany
| |
Collapse
|
26
|
Getmantseva L, Kolosova M, Bakoev F, Zimina A, Bakoev S. Genomic Regions and Candidate Genes Linked to Capped Hock in Pig. Life (Basel) 2021; 11:life11060510. [PMID: 34073088 PMCID: PMC8228005 DOI: 10.3390/life11060510] [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: 04/22/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 01/13/2023] Open
Abstract
Capped hock affects the exterior of pedigree pigs, making them unsalable and resulting in a negative impact on the efficiency of pig-breeding centers. The purpose of this paper was to carry out pilot studies aimed at finding genomic regions and genes linked to the capped hock in pigs. The studies were carried out on Landrace pigs (n = 75) and Duroc pigs (n = 70). To identify genomic regions linked to capped hock in pigs, we used smoothing FST statistics. Genotyping was performed with GeneSeek® GGP Porcine HD Genomic Profiler v1 (Illumina Inc, San Diego, CA, USA). The research results showed 70 SNPs linked to capped hock in Landrace (38 SNPs) and Duroc (32 SNPs). The identified regions overlapped with QTLs related with health traits (blood parameters) and meat and carcass traits (fatness). In total, 31 genes were identified (i.e., 17 genes in Landrace, 14 genes in Durocs). Three genes appeared in both the Landrace and Duroc groups, including A2ML1 (SSC5), ROBO2 (SSC13), and MSI1 (SSC14). We identified genomic regions directly or indirectly linked to capped hock, which thus might contribute to identifying genetic variants and using them as genetic markers in pig breeding.
Collapse
Affiliation(s)
- Lyubov Getmantseva
- Federal Research Center for Animal Husbandry Named after Academy Member L.K. Ernst, 142132 Dubrovitsy, Russia; (M.K.); (F.B.); (S.B.)
- Correspondence: (L.G.); (A.Z.); Tel.: +7-(4967)-65-11-01 (L.G. & A.Z.)
| | - Maria Kolosova
- Federal Research Center for Animal Husbandry Named after Academy Member L.K. Ernst, 142132 Dubrovitsy, Russia; (M.K.); (F.B.); (S.B.)
- Department of Biotechnology, Don State Agrarian University, 346493 Persianovski, Russia
| | - Faridun Bakoev
- Federal Research Center for Animal Husbandry Named after Academy Member L.K. Ernst, 142132 Dubrovitsy, Russia; (M.K.); (F.B.); (S.B.)
| | - Anna Zimina
- Federal Research Center for Animal Husbandry Named after Academy Member L.K. Ernst, 142132 Dubrovitsy, Russia; (M.K.); (F.B.); (S.B.)
- Correspondence: (L.G.); (A.Z.); Tel.: +7-(4967)-65-11-01 (L.G. & A.Z.)
| | - Siroj Bakoev
- Federal Research Center for Animal Husbandry Named after Academy Member L.K. Ernst, 142132 Dubrovitsy, Russia; (M.K.); (F.B.); (S.B.)
- Centre for Strategic Planning and Management of Biomedical Health Risks, 123182 Moscow, Russia
| |
Collapse
|
27
|
Kinoshita C, Kubota N, Aoyama K. Interplay of RNA-Binding Proteins and microRNAs in Neurodegenerative Diseases. Int J Mol Sci 2021; 22:ijms22105292. [PMID: 34069857 PMCID: PMC8157344 DOI: 10.3390/ijms22105292] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/14/2021] [Accepted: 05/15/2021] [Indexed: 02/07/2023] Open
Abstract
The number of patients with neurodegenerative diseases (NDs) is increasing, along with the growing number of older adults. This escalation threatens to create a medical and social crisis. NDs include a large spectrum of heterogeneous and multifactorial pathologies, such as amyotrophic lateral sclerosis, frontotemporal dementia, Alzheimer’s disease, Parkinson’s disease, Huntington’s disease and multiple system atrophy, and the formation of inclusion bodies resulting from protein misfolding and aggregation is a hallmark of these disorders. The proteinaceous components of the pathological inclusions include several RNA-binding proteins (RBPs), which play important roles in splicing, stability, transcription and translation. In addition, RBPs were shown to play a critical role in regulating miRNA biogenesis and metabolism. The dysfunction of both RBPs and miRNAs is often observed in several NDs. Thus, the data about the interplay among RBPs and miRNAs and their cooperation in brain functions would be important to know for better understanding NDs and the development of effective therapeutics. In this review, we focused on the connection between miRNAs, RBPs and neurodegenerative diseases.
Collapse
Affiliation(s)
- Chisato Kinoshita
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan;
- Correspondence: (C.K.); (K.A.); Tel.: +81-3-3964-3794 (C.K.); +81-3-3964-3793 (K.A.)
| | - Noriko Kubota
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan;
- Teikyo University Support Center for Women Physicians and Researchers, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Koji Aoyama
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan;
- Correspondence: (C.K.); (K.A.); Tel.: +81-3-3964-3794 (C.K.); +81-3-3964-3793 (K.A.)
| |
Collapse
|
28
|
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
|
29
|
Sun J, Wang X, Zhang Z, Zeng Z, Ouyang S, Kang W. The Sensitivity Prediction of Neoadjuvant Chemotherapy for Gastric Cancer. Front Oncol 2021; 11:641304. [PMID: 33937042 PMCID: PMC8085495 DOI: 10.3389/fonc.2021.641304] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/22/2021] [Indexed: 12/24/2022] Open
Abstract
The overall efficacy of neoadjuvant chemoradiotherapy (NACT) for locally advanced gastric cancer (LAGC) has been recognized. However, the response rate of NACT is limited due to tumor heterogeneity. For patients who are resistant to NACT, not only the operation timing will be postponed, patients will also suffer from the side effects of it. Thus, it is important to develop a comprehensive strategy and screen out patients who may be sensitive to NACT. This article summarizes the related research progress on the sensitivity prediction of NACT for GC in the following aspects: microRNAs, metabolic enzymes, exosomes, other biomarkers; inflammatory indicators, and imageological assessments. The results showed that there were many studies on biomarkers, but no unified conclusion has been drawn. The inflammatory indicators are related to the survival and prognosis of patients under NACT. For imageological assessments such as CT, MRI, and PET, with careful integration and optimization, they will have unique advantages in early screening for patients who are sensitive to NACT.
Collapse
Affiliation(s)
- Juan Sun
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Peking Union Medical College Hospital (CAMS), Beijing, China
| | - Xianze Wang
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Peking Union Medical College Hospital (CAMS), Beijing, China
| | - Zimu Zhang
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Peking Union Medical College Hospital (CAMS), Beijing, China
| | - Ziyang Zeng
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Peking Union Medical College Hospital (CAMS), Beijing, China
| | - Siwen Ouyang
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Peking Union Medical College Hospital (CAMS), Beijing, China
| | - Weiming Kang
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of General Surgery, Peking Union Medical College Hospital (CAMS), Beijing, China
| |
Collapse
|
30
|
Palacios F, Yan XJ, Ferrer G, Chen SS, Vergani S, Yang X, Gardner J, Barrientos JC, Rock P, Burack R, Kolitz JE, Allen SL, Kharas MG, Abdel-Wahab O, Rai KR, Chiorazzi N. Musashi 2 influences chronic lymphocytic leukemia cell survival and growth making it a potential therapeutic target. Leukemia 2021; 35:1037-1052. [PMID: 33504942 PMCID: PMC8024198 DOI: 10.1038/s41375-020-01115-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 11/04/2020] [Accepted: 12/14/2020] [Indexed: 01/30/2023]
Abstract
Progression of chronic lymphocytic leukemia (CLL) results from the expansion of a small fraction of proliferating leukemic B cells. When comparing the global gene expression of recently divided CLL cells with that of previously divided cells, we found higher levels of genes involved in regulating gene expression. One of these was the oncogene Musashi 2 (MSI2), an RNA-binding protein that induces or represses translation. While there is an established role for MSI2 in normal and malignant stem cells, much less is known about its expression and role in CLL. Here we report for the first time ex vivo and in vitro experiments that MSI2 protein levels are higher in dividing and recently divided leukemic cells and that downregulating MSI2 expression or blocking its function eliminates primary human and murine CLL and mature myeloid cells. Notably, mature T cells and hematopoietic stem and progenitor cells are not affected. We also confirm that higher MSI2 levels correlate with poor outcome markers, shorter time-to-first-treatment, and overall survival. Thus, our data highlight an important role for MSI2 in CLL-cell survival and proliferation and associate MSI2 with poor prognosis in CLL patients. Collectively, these findings pinpoint MSI2 as a potentially valuable therapeutic target in CLL.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents
- Apoptosis/drug effects
- Biomarkers, Tumor
- Caspase 3/metabolism
- Cell Cycle Checkpoints/drug effects
- Cell Line, Tumor
- Cell Survival/genetics
- Cyclin-Dependent Kinase Inhibitor p27/metabolism
- Disease Models, Animal
- Gene Expression
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic
- Gene Knockdown Techniques
- Humans
- Immunophenotyping
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Mice
- Molecular Targeted Therapy
- Prognosis
- RNA, Small Interfering
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Tumor Suppressor Protein p53/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Florencia Palacios
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Xiao-Jie Yan
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Gerardo Ferrer
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Shih-Shih Chen
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Stefano Vergani
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Xuejing Yang
- Molecular Pharmacology Program, Center for Cell Engineering, Center for Stem Cell Biology, Center for Experimental Therapeutics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jeffrey Gardner
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jaqueline C Barrientos
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Department of Medicine, Northwell Health, Manhasset and New Hyde Park, New York, NY, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Philip Rock
- Department of Pathology, University of Rochester, Rochester, NY, USA
| | - Richard Burack
- Department of Pathology, University of Rochester, Rochester, NY, USA
| | - Jonathan E Kolitz
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Department of Medicine, Northwell Health, Manhasset and New Hyde Park, New York, NY, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Steven L Allen
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Department of Medicine, Northwell Health, Manhasset and New Hyde Park, New York, NY, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Michael G Kharas
- Molecular Pharmacology Program, Center for Cell Engineering, Center for Stem Cell Biology, Center for Experimental Therapeutics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kanti R Rai
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Department of Medicine, Northwell Health, Manhasset and New Hyde Park, New York, NY, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Nicholas Chiorazzi
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA.
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Northwell Health, Manhasset and New Hyde Park, New York, NY, USA.
| |
Collapse
|
31
|
α1,3-Fucosyltransferase-IX, an enzyme of pulmonary endogenous lung stem cell marker SSEA-1, alleviates experimental bronchopulmonary dysplasia. Pediatr Res 2021; 89:1126-1135. [PMID: 32303051 DOI: 10.1038/s41390-020-0891-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 02/19/2020] [Accepted: 03/19/2020] [Indexed: 11/08/2022]
Abstract
BACKGROUND Endogenous pulmonary stem cells (PSCs) play an important role in lung development and repair; however, little is known about their role in bronchopulmonary dysplasia (BPD). We hypothesize that an endogenous PSC marker stage-specific embryonic antigen-1 (SSEA-1) and its enzyme, α1,3-fucosyltransferase IX (FUT9) play an important role in decreasing inflammation and restoring lung structure in experimental BPD. METHODS We studied the expression of SSEA-1, and its enzyme FUT9, in wild-type (WT) C57BL/6 mice, in room air and hyperoxia. Effects of intraperitoneal administration of recombinant human FUT9 (rhFUT9) on lung airway and parenchymal inflammation, alveolarization, and apoptosis were evaluated. RESULTS On hyperoxia exposure, SSEA-1 significantly decreased at postnatal day 14 in hyperoxia-exposed BPD mice, accompanied by a decrease in FUT9. BPD and respiratory distress syndrome (RDS) in human lungs showed decreased expression of SSEA-1 as compared to their term controls. Importantly, intraperitoneal administration of FUT9 in the neonatal BPD mouse model resulted in significant decrease in pulmonary airway (but not lung parenchymal) inflammation, alveolar-capillary leakage, alveolar simplification, and cell death in the hyperoxia-exposed BPD mice. CONCLUSIONS An important role of endogenous PSC marker SSEA-1 and its enzyme FUT9 is demonstrated, indicating early systemic intervention with FUT9 as a potential therapeutic option for BPD. IMPACT Administration of rhFUT9, an enzyme of endogenous stem cell marker SSEA-1, reduces pulmonary airway (but not lung parenchymal) inflammation, alveolar-capillary leak and cell death in the BPD mouse model. SSEA-1 is reported for the first time in experimental BPD models, and in human RDS and BPD. rhFUT9 treatment ameliorates hyperoxia-induced lung injury in a developmentally appropriate BPD mouse model. Our results have translational potential as a therapeutic modality for BPD in the developing lung.
Collapse
|
32
|
Padial-Molina M, Crespo-Lora V, Candido-Corral C, Martin-Morales N, Abril-Garcia D, Galindo-Moreno P, Hernandez-Cortes P, O’Valle F. Expression of Musashi-1 Increases in Bone Healing. Int J Mol Sci 2021; 22:ijms22073395. [PMID: 33810326 PMCID: PMC8037090 DOI: 10.3390/ijms22073395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/16/2022] Open
Abstract
Musashi-1 (MSI1) is an RNA-binding protein that regulates progenitor cells in adult and developing organisms to maintain self-renewal capacities. The role of musashi-1 in the bone healing environment and its relation with other osteogenic factors is unknown. In the current study, we analyze the expression of MSI1 in an experimental model of rat femoral bone fractures. We also analyze the relation between MSI1 expression and the expression of two osteogenic markers: periostin (POSTN) and runt-related transcription factor 2 (RUNX2). We use histological, immunohistochemical, and qPCR techniques to evaluate bone healing and the expression of MSI1, POSTN, and RUNX2 over time (4, 7, and 14 days). We compare our findings with non-fractured controls. We find that in bone calluses, the number of cells expressing MSI1 and RUNX2 increase over time and the intensity of POSTN expression decreases over time. Within bone calluses, we find the presence of MSI1 expression in mesenchymal stromal cells, osteoblasts, and osteocytes but not in hypertrophic chondrocytes. After 14 days, the expression of MSI1, POSTN, and RUNX2 was significantly correlated. Thus, we conclude that musashi-1 potentially serves in the osteogenic differentiation of mesenchymal stromal cells and bone healing. Therefore, further studies are needed to determine the possibility of musashi-1′s role as a clinical biomarker of bone healing and therapeutic agent for bone regeneration.
Collapse
Affiliation(s)
- Miguel Padial-Molina
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, and Centre for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (M.P.-M.); (N.M.-M.); (D.A.-G.)
| | - Vicente Crespo-Lora
- Department of Pathology, University of Granada, 18071 Granada, Spain; (V.C.-L.); (C.C.-C.)
| | - Clara Candido-Corral
- Department of Pathology, University of Granada, 18071 Granada, Spain; (V.C.-L.); (C.C.-C.)
| | - Nati Martin-Morales
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, and Centre for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (M.P.-M.); (N.M.-M.); (D.A.-G.)
- Department of Pathology, University of Granada, 18071 Granada, Spain; (V.C.-L.); (C.C.-C.)
| | - Dario Abril-Garcia
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, and Centre for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (M.P.-M.); (N.M.-M.); (D.A.-G.)
| | - Pablo Galindo-Moreno
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, and Centre for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (M.P.-M.); (N.M.-M.); (D.A.-G.)
- Correspondence:
| | - Pedro Hernandez-Cortes
- Department of Orthopedic Surgery, San Cecilio University Hospital, 18071 Granada, Spain;
| | - Francisco O’Valle
- Department of Pathology, Institute of Biopathology and Regenerative Medicine (IBIMER, CIBM), and Institute of Biosanitary (ibs-Granada), University of Granada, 18071 Granada, Spain;
| |
Collapse
|
33
|
Baroni M, Yi C, Choudhary S, Lei X, Kosti A, Grieshober D, Velasco M, Qiao M, Burns SS, Araujo PR, DeLambre T, Son MY, Plateroti M, Ferreira MAR, Hasty EP, Penalva LOF. Musashi1 Contribution to Glioblastoma Development via Regulation of a Network of DNA Replication, Cell Cycle and Division Genes. Cancers (Basel) 2021; 13:1494. [PMID: 33804958 PMCID: PMC8036803 DOI: 10.3390/cancers13071494] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/17/2021] [Accepted: 03/21/2021] [Indexed: 11/21/2022] Open
Abstract
RNA-binding proteins (RBPs) function as master regulators of gene expression. Alterations in their levels are often observed in tumors with numerous oncogenic RBPs identified in recent years. Musashi1 (Msi1) is an RBP and stem cell gene that controls the balance between self-renewal and differentiation. High Msi1 levels have been observed in multiple tumors including glioblastoma and are often associated with poor patient outcomes and tumor growth. A comprehensive genomic analysis identified a network of cell cycle/division and DNA replication genes and established these processes as Msi1's core regulatory functions in glioblastoma. Msi1 controls this gene network via two mechanisms: direct interaction and indirect regulation mediated by the transcription factors E2F2 and E2F8. Moreover, glioblastoma lines with Msi1 knockout (KO) displayed increased sensitivity to cell cycle and DNA replication inhibitors. Our results suggest that a drug combination strategy (Msi1 + cell cycle/DNA replication inhibitors) could be a viable route to treat glioblastoma.
Collapse
Affiliation(s)
- Mirella Baroni
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
| | - Caihong Yi
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
- Third Xiangya Hospital, Central South University, Changsha 410000, China
| | - Saket Choudhary
- Computational Biology and Bioinformatics, University of Southern California, Los Angeles, CA 90089, USA;
| | - Xiufen Lei
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
| | - Adam Kosti
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Denise Grieshober
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
| | - Mitzli Velasco
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
| | - Mei Qiao
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
| | - Suzanne S. Burns
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
| | - Patricia R. Araujo
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
| | - Talia DeLambre
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
| | - Mi Young Son
- Department of Molecular Medicine, Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX 78229, USA; (M.Y.S.); (E.P.H.)
| | - Michelina Plateroti
- Team: Development, Cancer and Stem Cells, Université de Strasbourg, Inserm, IRFAC/UMR-S1113, FMTS, 67200 Strasbourg, France;
| | | | - E. Paul Hasty
- Department of Molecular Medicine, Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX 78229, USA; (M.Y.S.); (E.P.H.)
| | - Luiz O. F. Penalva
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
34
|
Yuan P, Ling L, Gao X, Sun T, Miao J, Yuan X, Liu J, Wang Z, Liu B. Identification of RNA-binding protein SNRPA1 for prognosis in prostate cancer. Aging (Albany NY) 2021; 13:2895-2911. [PMID: 33460399 PMCID: PMC7880319 DOI: 10.18632/aging.202387] [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] [Received: 06/03/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023]
Abstract
Prostate cancer is one of the deadliest cancers in men. RNA-binding proteins play a critical role in human cancers; however, whether they have a significant effect on the prognosis of prostate cancer has yet to be elucidated. In the present study, we performed a comprehensive analysis of RNA sequencing and clinical data from the Cancer Genome Atlas dataset and obtained differentially expressed RNA-binding proteins between prostate cancer and benign tissues. We constructed a protein-protein interaction network and Cox regression analyses were conducted to identify prognostic hub RNA-binding proteins. SNRPA1 was associated with the highest risk of poor prognosis and was therefore selected for further analysis. SNRPA1 expression was positively correlated with Gleason score and pathological TNM stage in prostate cancer patients. Furthermore, the expression profile of SNRPA1 was validated using the Oncomine, Human Protein Atlas, and Cancer Cell Line Encyclopedia databases. Meanwhile, the prognostic profile of SNRPA1 was successfully verified in GSE70769. Additionally, the results of molecular experiments revealed the proliferative role of SNRPA1 in prostate cancer cells. In summary, our findings evidenced a relationship between RNA-binding proteins and prostate cancer and indicated the prognostic significance of SNRPA1 in prostate cancer.
Collapse
Affiliation(s)
- Penghui Yuan
- Department of Urology Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Le Ling
- Department of Urology Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Xintao Gao
- Department of Urology Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Taotao Sun
- Department of Urology Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Jianping Miao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Jihong Liu
- Department of Urology Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Zhihua Wang
- Department of Urology Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| |
Collapse
|
35
|
Orzechowska EJ, Katano T, Bialkowska AB, Yang VW. Interplay among p21 Waf1/Cip1, MUSASHI-1 and Krüppel-like factor 4 in activation of Bmi1-Cre ER reserve intestinal stem cells after gamma radiation-induced injury. Sci Rep 2020; 10:18300. [PMID: 33110120 PMCID: PMC7591575 DOI: 10.1038/s41598-020-75171-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 10/12/2020] [Indexed: 12/23/2022] Open
Abstract
Gamma radiation is a commonly used adjuvant treatment for abdominally localized cancer. Since its therapeutic potential is limited due to gastrointestinal (GI) syndrome, elucidation of the regenerative response following radiation-induced gut injury is needed to develop a preventive treatment. Previously, we showed that Krüppel-like factor 4 (KLF4) activates certain quiescent intestinal stem cells (ISCs) marked by Bmi1-CreER to give rise to regenerating crypts following γ irradiation. In the current study, we showed that γ radiation-induced expression of p21Waf1/Cip1 in Bmi1-CreER cells is likely mitigated by MUSASHI-1 (MSI1) acting as a negative regulator of p21Waf1/Cip1 mRNA translation, which promotes exit of the Bmi1-CreER cells from a quiescent state. Additionally, Bmi1-specific Klf4 deletion resulted in decreased numbers of MSI1+ cells in regenerating crypts compared to those of control mice. We showed that KLF4 binds to the Msi1 promoter and activates its expression in vitro. Since MSI1 has been shown to be crucial for crypt regeneration, this finding elucidates a pro-proliferative role of KLF4 during the postirradiation regenerative response. Taken together, our data suggest that the interplay among p21Waf1/Cip1, MSI1 and KLF4 regulates Bmi1-CreER cell survival, exit from quiescence and regenerative potential upon γ radiation-induced injury.
Collapse
Affiliation(s)
- Emilia J Orzechowska
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.,Department of Molecular Biology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Takahito Katano
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.,Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Agnieszka B Bialkowska
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.
| | - Vincent W Yang
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA. .,Department of Physiology and Biophysics, Renaissance School of Medicine at Stony, Brook University, Stony Brook, NY, USA.
| |
Collapse
|
36
|
Naef V, De Sarlo M, Testa G, Corsinovi D, Azzarelli R, Borello U, Ori M. The Stemness Gene Mex3A Is a Key Regulator of Neuroblast Proliferation During Neurogenesis. Front Cell Dev Biol 2020; 8:549533. [PMID: 33072742 PMCID: PMC7536324 DOI: 10.3389/fcell.2020.549533] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 08/31/2020] [Indexed: 01/31/2023] Open
Abstract
Mex3A is an RNA binding protein that can also act as an E3 ubiquitin ligase to control gene expression at the post-transcriptional level. In intestinal adult stem cells, MEX3A is required for cell self-renewal and when overexpressed, MEX3A can contribute to support the proliferation of different cancer cell types. In a completely different context, we found mex3A among the genes expressed in neurogenic niches of the embryonic and adult fish brain and, notably, its expression was downregulated during brain aging. The role of mex3A during embryonic and adult neurogenesis in tetrapods is still unknown. Here, we showed that mex3A is expressed in the proliferative region of the developing brain in both Xenopus and mouse embryos. Using gain and loss of gene function approaches, we showed that, in Xenopus embryos, mex3A is required for neuroblast proliferation and its depletion reduced the neuroblast pool, leading to microcephaly. The tissue-specific overexpression of mex3A in the developing neural plate enhanced the expression of sox2 and msi-1 keeping neuroblasts into a proliferative state. It is now clear that the stemness property of mex3A, already demonstrated in adult intestinal stem cells and cancer cells, is a key feature of mex3a also in developing brain, opening new lines of investigation to better understand its role during brain aging and brain cancer development.
Collapse
Affiliation(s)
- Valentina Naef
- Unità di Biologia Cellulare e dello Sviluppo, Dipartimento di Biologia, Università di Pisa, Pisa, Italy.,Molecular Medicine, IRCCS Fondazione Stella Maris, Pisa, Italy
| | - Miriam De Sarlo
- Unità di Biologia Cellulare e dello Sviluppo, Dipartimento di Biologia, Università di Pisa, Pisa, Italy
| | - Giovanna Testa
- Unità di Biologia Cellulare e dello Sviluppo, Dipartimento di Biologia, Università di Pisa, Pisa, Italy.,Scuola Normale Superiore di Pisa, Pisa, Italy
| | - Debora Corsinovi
- Unità di Biologia Cellulare e dello Sviluppo, Dipartimento di Biologia, Università di Pisa, Pisa, Italy
| | - Roberta Azzarelli
- Unità di Biologia Cellulare e dello Sviluppo, Dipartimento di Biologia, Università di Pisa, Pisa, Italy
| | - Ugo Borello
- Unità di Biologia Cellulare e dello Sviluppo, Dipartimento di Biologia, Università di Pisa, Pisa, Italy
| | - Michela Ori
- Unità di Biologia Cellulare e dello Sviluppo, Dipartimento di Biologia, Università di Pisa, Pisa, Italy
| |
Collapse
|
37
|
Evolutionary analysis of the Musashi family: What can it tell us about Zika? INFECTION GENETICS AND EVOLUTION 2020; 84:104364. [DOI: 10.1016/j.meegid.2020.104364] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 12/21/2022]
|
38
|
Mendonça LS, Nóbrega C, Tavino S, Brinkhaus M, Matos C, Tomé S, Moreira R, Henriques D, Kaspar BK, Pereira de Almeida L. Ibuprofen enhances synaptic function and neural progenitors proliferation markers and improves neuropathology and motor coordination in Machado-Joseph disease models. Hum Mol Genet 2020; 28:3691-3703. [PMID: 31127937 DOI: 10.1093/hmg/ddz097] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 04/22/2019] [Accepted: 05/07/2019] [Indexed: 01/09/2023] Open
Abstract
Machado-Joseph disease or spinocerebellar ataxia type 3 is an inherited neurodegenerative disease associated with an abnormal glutamine over-repetition within the ataxin-3 protein. This mutant ataxin-3 protein affects several cellular pathways, leading to neuroinflammation and neuronal death in specific brain regions resulting in severe clinical manifestations. Presently, there is no therapy able to modify the disease progression. Nevertheless, anti-inflammatory pharmacological intervention has been associated with positive outcomes in other neurodegenerative diseases. Thus, the present work aimed at investigating whether ibuprofen treatment would alleviate Machado-Joseph disease. We found that ibuprofen-treated mouse models presented a significant reduction in the neuroinflammation markers, namely Il1b and TNFa mRNA and IKB-α protein phosphorylation levels. Moreover, these mice exhibited neuronal preservation, cerebellar atrophy reduction, smaller mutant ataxin-3 inclusions and motor performance improvement. Additionally, neural cultures of Machado-Joseph disease patients' induced pluripotent stem cells-derived neural stem cells incubated with ibuprofen showed increased levels of neural progenitors proliferation and synaptic markers such as MSI1, NOTCH1 and SYP. These findings were further confirmed in ibuprofen-treated mice that display increased neural progenitor numbers (Ki67 positive) in the subventricular zone. Furthermore, interestingly, ibuprofen treatment enhanced neurite total length and synaptic function of human neurons. Therefore, our results indicate that ibuprofen reduces neuroinflammation and induces neuroprotection, alleviating Machado-Joseph disease-associated neuropathology and motor impairments. Thus, our findings demonstrate that ibuprofen treatment has the potential to be used as a neuroprotective therapeutic approach in Machado-Joseph disease.
Collapse
Affiliation(s)
- Liliana S Mendonça
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Clévio Nóbrega
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Silvia Tavino
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Maximilian Brinkhaus
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Carlos Matos
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Sandra Tomé
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ricardo Moreira
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Daniel Henriques
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Brian K Kaspar
- The Research Institute at Nationwide Children's Hospital, Ohio State University School of Medicine, Columbus, Ohio 43205, USA
| | - Luís Pereira de Almeida
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
39
|
Knockdown of Musashi RNA Binding Proteins Decreases Radioresistance but Enhances Cell Motility and Invasion in Triple-Negative Breast Cancer. Int J Mol Sci 2020; 21:ijms21062169. [PMID: 32245259 PMCID: PMC7139790 DOI: 10.3390/ijms21062169] [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: 12/29/2019] [Revised: 03/19/2020] [Accepted: 03/19/2020] [Indexed: 12/28/2022] Open
Abstract
The therapeutic potential of Musashi (MSI) RNA-binding proteins, important stemness-associated gene expression regulators, remains insufficiently understood in breast cancer. This study identifies the interplay between MSI protein expression, stem cell characteristics, radioresistance, cell invasiveness and migration. MSI-1, MSI-2 and Notch pathway elements were investigated via quantitative polymerase chain reaction (qPCR) in 19 triple-negative breast cancer samples. Measurements were repeated in MDA-MB-231 cells after MSI-1 and -2 siRNA-mediated double knockdown, with further experiments performed after MSI silencing. Flow cytometry helped quantify expression of CD44 and leukemia inhibitory factor receptor (LIFR), changes in apoptosis and cell cycle progression. Proliferation and irradiation-induced effects were assessed using colony formation assays. Radiation-related proteins were investigated via Western blots. Finally, cell invasion assays and digital holographic microscopy for cell migration were performed. MSI proteins showed strong correlations with Notch pathway elements. MSI knockdown resulted in reduction of stem cell marker expression, cell cycle progression and proliferation, while increasing apoptosis. Cells were radiosensitized as radioresistance-conferring proteins were downregulated. However, MSI-silencing-mediated LIFR downregulation resulted in enhanced cell invasion and migration. We conclude that, while MSI knockdown results in several therapeutically desirable consequences, enhanced invasion and migration need to be counteracted before knockdown advantages can be fully exploited.
Collapse
|
40
|
Wang X, Voronina E. Diverse Roles of PUF Proteins in Germline Stem and Progenitor Cell Development in C. elegans. Front Cell Dev Biol 2020; 8:29. [PMID: 32117964 PMCID: PMC7015873 DOI: 10.3389/fcell.2020.00029] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 01/14/2020] [Indexed: 01/05/2023] Open
Abstract
Stem cell development depends on post-transcriptional regulation mediated by RNA-binding proteins (RBPs) (Zhang et al., 1997; Forbes and Lehmann, 1998; Okano et al., 2005; Ratti et al., 2006; Kwon et al., 2013). Pumilio and FBF (PUF) family RBPs are highly conserved post-transcriptional regulators that are critical for stem cell maintenance (Wickens et al., 2002; Quenault et al., 2011). The RNA-binding domains of PUF proteins recognize a family of related sequence motifs in the target mRNAs, yet individual PUF proteins have clearly distinct biological functions (Lu et al., 2009; Wang et al., 2018). The C. elegans germline is a simple and powerful model system for analyzing regulation of stem cell development. Studies in C. elegans uncovered specific physiological roles for PUFs expressed in the germline stem cells ranging from control of proliferation and differentiation to regulation of the sperm/oocyte decision. Importantly, recent studies started to illuminate the mechanisms behind PUF functional divergence. This review summarizes the many roles of PUF-8, FBF-1, and FBF-2 in germline stem and progenitor cells (SPCs) and discusses the factors accounting for their distinct biological functions. PUF proteins are conserved in evolution, and insights into PUF-mediated regulation provided by the C. elegans model system are likely relevant for other organisms.
Collapse
Affiliation(s)
- Xiaobo Wang
- Division of Biological Sciences, University of Montana, Missoula, MT, United States
| | - Ekaterina Voronina
- Division of Biological Sciences, University of Montana, Missoula, MT, United States
| |
Collapse
|
41
|
Urooj T, Wasim B, Mushtaq S, Shah SNN, Shah M. Cancer Cell-derived Secretory Factors in Breast Cancer-associated Lung Metastasis: Their Mechanism and Future Prospects. Curr Cancer Drug Targets 2020; 20:168-186. [PMID: 31858911 PMCID: PMC7516334 DOI: 10.2174/1568009620666191220151856] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/09/2019] [Accepted: 12/18/2019] [Indexed: 02/07/2023]
Abstract
In Breast cancer, Lung is the second most common site of metastasis after the bone. Various factors are responsible for Lung metastasis occurring secondary to Breast cancer. Cancer cellderived secretory factors are commonly known as 'Cancer Secretomes'. They exhibit a prompt role in the mechanism of Breast cancer lung metastasis. They are also major constituents of hostassociated tumor microenvironment. Through cross-talk between cancer cells and the extracellular matrix components, cancer cell-derived extracellular matrix components (CCECs) such as hyaluronan, collagens, laminin and fibronectin cause ECM remodeling at the primary site (breast) of cancer. However, at the secondary site (lung), tenascin C, periostin and lysyl oxidase, along with pro-metastatic molecules Coco and GALNT14, contribute to the formation of pre-metastatic niche (PMN) by promoting ECM remodeling and lung metastatic cells colonization. Cancer cell-derived secretory factors by inducing cancer cell proliferation at the primary site, their invasion through the tissues and vessels and early colonization of metastatic cells in the PMN, potentiate the mechanism of Lung metastasis in Breast cancer. On the basis of biochemical structure, these secretory factors are broadly classified into proteins and non-proteins. This is the first review that has highlighted the role of cancer cell-derived secretory factors in Breast cancer Lung metastasis (BCLM). It also enumerates various researches that have been conducted to date in breast cancer cell lines and animal models that depict the prompt role of various types of cancer cell-derived secretory factors involved in the process of Breast cancer lung metastasis. In the future, by therapeutically targeting these cancer driven molecules, this specific type of organ-tropic metastasis in breast cancer can be successfully treated.
Collapse
Affiliation(s)
- Tabinda Urooj
- Anatomy Department, Ziauddin University, Clifton Karachi, Sindh, Pakistan
| | - Bushra Wasim
- Anatomy Department, Ziauddin University, Clifton Karachi, Sindh, Pakistan
| | - Shamim Mushtaq
- Biochemistry Department, Ziauddin University, Clifton Karachi, Sindh, Pakistan
| | | | - Muzna Shah
- Anatomy Department, Ziauddin University, Clifton Karachi, Sindh, Pakistan
| |
Collapse
|
42
|
Maheshvara, a Conserved RNA Helicase, Regulates Notch Signaling in Drosophila melanogaster. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1227:69-79. [DOI: 10.1007/978-3-030-36422-9_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
43
|
Montalbano M, McAllen S, Sengupta U, Puangmalai N, Bhatt N, Ellsworth A, Kayed R. Tau oligomers mediate aggregation of RNA-binding proteins Musashi1 and Musashi2 inducing Lamin alteration. Aging Cell 2019; 18:e13035. [PMID: 31532069 PMCID: PMC6826126 DOI: 10.1111/acel.13035] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 08/07/2019] [Accepted: 08/17/2019] [Indexed: 01/01/2023] Open
Abstract
The exact mechanisms leading to neurodegeneration in Alzheimer's disease (AD) and other tauopathies are not yet entirely understood. However, it is known that several RNA-binding proteins (RBPs) form toxic aggregates and also interact with tau in such granules in tauopathies, including AD. The Musashi (MSI) family of RBPs, consisting of two homologues: Musashi1 and Musashi2, have not been extensively investigated in neurodegenerative diseases. Here, using a tau inducible HEK (iHEK) model we investigate whether MSI proteins contribute to the aggregation of toxic tau oligomers (TauO). Wild-type and mutant P301L tau iHEK cells are used to study the effect of different tau variants on the cellular localization of MSI proteins. Interestingly, we observe that tau co-localizes with MSI in the cytoplasm and nuclei, altering the nuclear transport of MSI. Furthermore, incremental changes in the size and density of nuclear MSI/tau foci are observed. We also report here that TauO interact with MSI to cause the formation of distinct nuclear aggregates. Moreover, tau/MSI aggregates induce structural changes to LaminB1, leading to nuclear instability. These results illustrate a possible mechanism of neurodegeneration mediated by the aggregation of MSI proteins and TauO, suggesting that MSI plays a critical role in cellular dysfunction.
Collapse
Affiliation(s)
- Mauro Montalbano
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTXUSA
- Department of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTXUSA
| | - Salome McAllen
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTXUSA
- Department of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTXUSA
| | - Urmi Sengupta
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTXUSA
- Department of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTXUSA
| | - Nicha Puangmalai
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTXUSA
- Department of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTXUSA
| | - Nemil Bhatt
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTXUSA
- Department of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTXUSA
| | - Anna Ellsworth
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTXUSA
- Department of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTXUSA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTXUSA
- Department of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTXUSA
| |
Collapse
|
44
|
Kinoshita M, Fujimoto C, Iwasaki S, Kashio A, Kikkawa YS, Kondo K, Okano H, Yamasoba T. Alteration of Musashi1 Intra-cellular Distribution During Regeneration Following Gentamicin-Induced Hair Cell Loss in the Guinea Pig Crista Ampullaris. Front Cell Neurosci 2019; 13:481. [PMID: 31708751 PMCID: PMC6824208 DOI: 10.3389/fncel.2019.00481] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 10/10/2019] [Indexed: 11/13/2022] Open
Abstract
The mechanism underlying hair cell (HC) regeneration in the mammalian inner ear is still under debate. Understanding what molecules regulate the HC regeneration in mature mammals will be the key to the treatment of the inner ear disorder. Musashi1 (MSI1) is an RNA binding protein associated with asymmetric division and maintenance of stem cell function as a modulator of the Notch-1 signaling pathway. In this study, we investigated the cellular proliferative activity and changes in spatiotemporal pattern of MSI1 expression in the gentamicin (GM)-treated crista ampullaris (CA) in guinea pigs. Although the vestibular HCs in the CA almost disappeared at 14 days after injecting GM in the inner ear, the density of vestibular HCs spontaneously increased by up to 50% relative to controls at 56 days post-GM treatment (PT). The number of the type II HCs was significantly increased at 28 days PT relative to 14 days PT (p < 0.01) while that of type I HCs or supporting cells (SCs) did not change. The number of SCs did not change through the observational period. Administration of bromodeoxyuridine with the same GM treatment showed that the cell proliferation activity was high in SCs between 14 and 28 days PT. The changes in spatiotemporal patterns of MSI1 expression during spontaneous HC regeneration following GM treatment showed that MSI1-immunoreactivity was diffusely spread into the cytoplasm of the SCs during 7–21 days PT whereas the expression of MSI1 was confined to the nucleus of SCs in the other period. The MSI1/MYO7A double-positive cells were observed at 21 days PT. These results suggest that regeneration of vestibular HCs might originate in the asymmetric cell division and differentiation of SCs and that MSI1 might be involved in controlling the process of vestibular HC regeneration.
Collapse
Affiliation(s)
- Makoto Kinoshita
- Department of Otolaryngology and Head and Neck Surgery, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Chisato Fujimoto
- Department of Otolaryngology and Head and Neck Surgery, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Shinichi Iwasaki
- Department of Otolaryngology and Head and Neck Surgery, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Akinori Kashio
- Department of Otolaryngology and Head and Neck Surgery, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Yayoi S Kikkawa
- Department of Otolaryngology and Head and Neck Surgery, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Kenji Kondo
- Department of Otolaryngology and Head and Neck Surgery, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Tatsuya Yamasoba
- Department of Otolaryngology and Head and Neck Surgery, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| |
Collapse
|
45
|
Yang LY, Song GL, Zhai XQ, Wang L, Liu QL, Zhou MS. MicroRNA-331 inhibits development of gastric cancer through targeting musashi1. World J Gastrointest Oncol 2019; 11:705-716. [PMID: 31558975 PMCID: PMC6755110 DOI: 10.4251/wjgo.v11.i9.705] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/23/2019] [Accepted: 07/17/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The molecular mechanisms involved in microRNAs (miRNAs) have been extensively investigated in gastric cancer (GC). However, how miR-331 regulates GC pathogenesis remains unknown.
AIM To illuminate the effect of miR-331 on cell metastasis and tumor growth in GC.
METHODS The qRT-PCR, CCK8, Transwell, cell adhesion, Western blot, luciferase reporter and xenograft tumor formation assays were applied to explore the regulatory mechanism of miR-331 in GC.
RESULTS Downregulation of miR-331 associated with poor prognosis was detected in GC. Functionally, miR-331 suppressed cell proliferation, metastasis and tumor growth in GC. Further, miR-331 was verified to directly target musashi1 (MSI1). In addition, miR-331 inversely regulated MSI1 expression in GC tissues. Furthermore, upregulation of MSI1 weakened the inhibitory effect of miR-331 in GC.
CONCLUSION miR-331 inhibited development of GC through targeting MSI1, which may be used as an indicator for the prediction and prognosis of GC.
Collapse
Affiliation(s)
- Lei-Ying Yang
- Department of Pathology, Shandong First Medical University, Taian 271016, Shandong Province, China
| | - Guang-Le Song
- Morphological Laboratory, Shandong First Medical University, Taian 271016, Shandong Province, China
| | - Xiao-Qian Zhai
- Department of Pathology, Second Affiliated Hospital of Shandong First Medical University, Taian 271016, Shandong Province, China
| | - Li Wang
- Department of Pathology, Shandong First Medical University, Taian 271016, Shandong Province, China
| | - Qin-Lai Liu
- Department of Pathology, Shandong First Medical University, Taian 271016, Shandong Province, China
| | - Ming-Shun Zhou
- Department of Emergency, Second Affiliated Hospital of Shandong First Medical University, Taian 271016, Shandong Province, China
| |
Collapse
|
46
|
García-Cárdenas JM, Guerrero S, López-Cortés A, Armendáriz-Castillo I, Guevara-Ramírez P, Pérez-Villa A, Yumiceba V, Zambrano AK, Leone PE, Paz-y-Miño C. Post-transcriptional Regulation of Colorectal Cancer: A Focus on RNA-Binding Proteins. Front Mol Biosci 2019; 6:65. [PMID: 31440515 PMCID: PMC6693420 DOI: 10.3389/fmolb.2019.00065] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/23/2019] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is a major health problem with an estimated 1. 8 million new cases worldwide. To date, most CRC studies have focused on DNA-related aberrations, leaving post-transcriptional processes under-studied. However, post-transcriptional alterations have been shown to play a significant part in the maintenance of cancer features. RNA binding proteins (RBPs) are uprising as critical regulators of every cancer hallmark, yet little is known regarding the underlying mechanisms and key downstream oncogenic targets. Currently, more than a thousand RBPs have been discovered in humans and only a few have been implicated in the carcinogenic process and even much less in CRC. Identification of cancer-related RBPs is of great interest to better understand CRC biology and potentially unveil new targets for cancer therapy and prognostic biomarkers. In this work, we reviewed all RBPs which have a role in CRC, including their control by microRNAs, xenograft studies and their clinical implications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - César Paz-y-Miño
- Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigación Genética y Genómica, Universidad UTE, Quito, Ecuador
| |
Collapse
|
47
|
Cragle CE, MacNicol MC, Byrum SD, Hardy LL, Mackintosh SG, Richardson WA, Gray NK, Childs GV, Tackett AJ, MacNicol AM. Musashi interaction with poly(A)-binding protein is required for activation of target mRNA translation. J Biol Chem 2019; 294:10969-10986. [PMID: 31152063 PMCID: PMC6635449 DOI: 10.1074/jbc.ra119.007220] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 05/09/2019] [Indexed: 12/21/2022] Open
Abstract
The Musashi family of mRNA translational regulators controls both physiological and pathological stem cell self-renewal primarily by repressing target mRNAs that promote differentiation. In response to differentiation cues, Musashi can switch from a repressor to an activator of target mRNA translation. However, the molecular events that distinguish Musashi-mediated translational activation from repression are not understood. We have previously reported that Musashi function is required for the maturation of Xenopus oocytes and specifically for translational activation of specific dormant maternal mRNAs. Here, we employed MS to identify cellular factors necessary for Musashi-dependent mRNA translational activation. We report that Musashi1 needs to associate with the embryonic poly(A)-binding protein (ePABP) or the canonical somatic cell poly(A)-binding protein PABPC1 for activation of Musashi target mRNA translation. Co-immunoprecipitation studies demonstrated an increased Musashi1 interaction with ePABP during oocyte maturation. Attenuation of endogenous ePABP activity severely compromised Musashi function, preventing downstream signaling and blocking oocyte maturation. Ectopic expression of either ePABP or PABPC1 restored Musashi-dependent mRNA translational activation and maturation of ePABP-attenuated oocytes. Consistent with these Xenopus findings, PABPC1 remained associated with Musashi under conditions of Musashi target mRNA de-repression and translation during mammalian stem cell differentiation. Because association of Musashi1 with poly(A)-binding proteins has previously been implicated only in repression of Musashi target mRNAs, our findings reveal novel context-dependent roles for the interaction of Musashi with poly(A)-binding protein family members in response to extracellular cues that control cell fate.
Collapse
Affiliation(s)
- Chad E Cragle
- Department of Neurobiology and Developmental Sciences
| | - Melanie C MacNicol
- Department of Neurobiology and Developmental Sciences,; Center for Translational Neuroscience
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology,; Arkansas Children's Research Institute
| | - Linda L Hardy
- Department of Neurobiology and Developmental Sciences
| | | | - William A Richardson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - Nicola K Gray
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - Gwen V Childs
- Department of Neurobiology and Developmental Sciences,; Center for Translational Neuroscience
| | - Alan J Tackett
- Department of Biochemistry and Molecular Biology,; Arkansas Children's Research Institute
| | - Angus M MacNicol
- Department of Neurobiology and Developmental Sciences,; Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205 and.
| |
Collapse
|
48
|
Ahlfors JE, Azimi A, El-Ayoubi R, Velumian A, Vonderwalde I, Boscher C, Mihai O, Mani S, Samoilova M, Khazaei M, Fehlings MG, Morshead CM. Examining the fundamental biology of a novel population of directly reprogrammed human neural precursor cells. Stem Cell Res Ther 2019; 10:166. [PMID: 31196173 PMCID: PMC6567617 DOI: 10.1186/s13287-019-1255-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cell reprogramming is a promising avenue for cell-based therapies as it allows for the generation of multipotent, unipotent, or mature somatic cells without going through a pluripotent state. While the use of autologous cells is considered ideal, key challenges for their clinical translation include the ability to reproducibly generate sufficient quantities of cells within a therapeutically relevant time window. METHODS We performed transfection of three distinct human somatic starting populations of cells with a non-integrating synthetic plasmid expressing Musashi 1 (MSI1), Neurogenin 2 (NGN2), and Methyl-CpG-Binding Domain 2 (MBD2). The resulting directly reprogrammed neural precursor cells (drNPCs) were examined in vitro using RT-qPCR, karyotype analysis, immunohistochemistry, and FACS at early and late time post-transfection. Electrophysiology (patch clamp) was performed on drNPC-derived neurons to determine their capacity to generate action potentials. In vivo characterization was performed following transplantation of drNPCs into two animal models (Shiverer and SCID/Beige mice), and the numbers, location, and differentiation profile of the transplanted cells were examined using immunohistochemistry. RESULTS Human somatic cells can be directly reprogrammed within two weeks to neural precursor cells (drNPCs) by transient exposure to Msi1, Ngn2, and MBD2 using non-viral constructs. The drNPCs generate all three neural cell types (astrocytes, oligodendrocytes, and neurons) and can be passaged in vitro to generate large numbers of cells within four weeks. drNPCs can respond to in vivo differentiation and migration cues as demonstrated by their migration to the olfactory bulb and contribution to neurogenesis in vivo. Differentiation profiles of transplanted cells onto the corpus callosum of myelin-deficient mice reveal the production of oligodendrocytes and astrocytes. CONCLUSIONS Human drNPCs can be efficiently and rapidly produced from donor somatic cells and possess all the important characteristics of native neural multipotent cells including differentiation into neurons, astrocytes, and oligodendrocytes, and in vivo neurogenesis and myelination.
Collapse
Affiliation(s)
| | - Ashkan Azimi
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario M5S 1A8 Canada
- Division of Anatomy, Department of Surgery, University of Toronto, Ontario, M5S 1A8 Canada
| | | | - Alexander Velumian
- Division of Neurosurgery, Department of Surgery, University of Toronto, Ontario, M5T 1P5 Canada
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario M5T 2S8 Canada
| | - Ilan Vonderwalde
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9 Canada
| | | | - Oana Mihai
- New World Laboratories, Laval, Quebec, H7V 5B7 Canada
| | - Sarathi Mani
- New World Laboratories, Laval, Quebec, H7V 5B7 Canada
| | - Marina Samoilova
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario M5T 2S8 Canada
| | - Mohamad Khazaei
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario M5T 2S8 Canada
| | - Michael G. Fehlings
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario M5S 1A8 Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Ontario, M5T 1P5 Canada
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario M5T 2S8 Canada
| | - Cindi M Morshead
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario M5S 1A8 Canada
- Division of Anatomy, Department of Surgery, University of Toronto, Ontario, M5S 1A8 Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9 Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S3E1 Canada
| |
Collapse
|
49
|
Moradi F, Babashah S, Sadeghizadeh M, Jalili A, Hajifathali A, Roshandel H. Signaling pathways involved in chronic myeloid leukemia pathogenesis: The importance of targeting Musashi2-Numb signaling to eradicate leukemia stem cells. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:581-589. [PMID: 31231484 PMCID: PMC6570743 DOI: 10.22038/ijbms.2019.31879.7666] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 11/15/2018] [Indexed: 11/06/2022]
Abstract
OBJECTIVES Chronic myeloid leukemia (CML) is a myeloid clonal proliferation disease defining by the presence of the Philadelphia chromosome that shows the movement of BCR-ABL1. In this study, the critical role of the Musashi2-Numb axis in determining cell fate and relationship of the axis to important signaling pathways such as Hedgehog and Notch that are essential for self-renewal pathways in CML stem cells will be reviewed meticulously. MATERIALS AND METHODS In this review, a PubMed search using the keywords of Leukemia, signaling pathways, Musashi2-Numb was performed, and then we summarized different research works . RESULTS Although tyrosine kinase inhibitors such as Imatinib significantly kill and remove the cell with BCR-ABL1 translocation, they are unable to target BCR-ABL1 leukemia stem cells. The main problem is stem cells resistance to Imatinib therapy. Therefore, the identification and control of downstream molecules/ signaling route of the BCR-ABL1 that are involved in the survival and self-renewal of leukemia stem cells can be an effective treatment strategy to eliminate leukemia stem cells, which supposed to be cured by Musashi2-Numb signaling pathway. CONCLUSION The control of molecules /pathways downstream of the BCR-ABL1 and targeting Musashi2-Numb can be an effective therapeutic strategy for treatment of chronic leukemia stem cells. While Musashi2 is a poor prognostic marker in leukemia, in treatment and strategy, it has significant diagnostic value.
Collapse
Affiliation(s)
- Foruzan Moradi
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Majid Sadeghizadeh
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Arsalan Jalili
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Abbas Hajifathali
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hajifathali Roshandel
- Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Hematology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
50
|
Nucleoporin Nup155 is part of the p53 network in liver cancer. Nat Commun 2019; 10:2147. [PMID: 31089132 PMCID: PMC6517424 DOI: 10.1038/s41467-019-10133-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 04/16/2019] [Indexed: 02/08/2023] Open
Abstract
Cancer-relevant signalling pathways rely on bidirectional nucleocytoplasmic transport events through the nuclear pore complex (NPC). However, mechanisms by which individual NPC components (Nups) participate in the regulation of these pathways remain poorly understood. We discover by integrating large scale proteomics, polysome fractionation and a focused RNAi approach that Nup155 controls mRNA translation of p21 (CDKN1A), a key mediator of the p53 response. The underlying mechanism involves transcriptional regulation of the putative tRNA and rRNA methyltransferase FTSJ1 by Nup155. Furthermore, we observe that Nup155 and FTSJ1 are p53 repression targets and accordingly find a correlation between the p53 status, Nup155 and FTSJ1 expression in murine and human hepatocellular carcinoma. Our data suggest an unanticipated regulatory network linking translational control by and repression of a structural NPC component modulating the p53 pathway through its effectors. The nuclear pore complex (NPC) is known to regulate p53 signaling and this has mainly been linked to peripheral NPC subunits. Here the authors show that Nup155 from the NPC inner ring regulates the p53 pathway by controlling p21 translation while also being a target of p53-mediated repression.
Collapse
|