1
|
Richard SA. The pivotal role of autophagy in the pathogenesis and therapy of medulloblastoma. Future Oncol 2024; 20:3313-3324. [PMID: 39513232 PMCID: PMC11633412 DOI: 10.1080/14796694.2024.2420629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 10/21/2024] [Indexed: 11/15/2024] Open
Abstract
Medulloblastoma (MB) is the most frequent malignant brain tumor in children. MB originates from neural precursor cells in distinctive regions of the rhombic lip and their maturation occurs in the cerebellum or the brain stem during embryonal development. Autophagy is also referred to as self-eating' which is a catabolic process that often triggers cellular homeostasis through the salvaging of degenerated proteins as well as organelles. Autophagy influence cell survival via aberrant proteins that could accumulate within the cell and influence potential signaling and transport mechanisms. The role of autophagy in MB aggressiveness as well as tumorigenesis is a very complex process. This review targets specifically data reporting the key roles of autophagy in the pathogenesis and therapy of MB.
Collapse
Affiliation(s)
- Seidu A. Richard
- Department of Biochemistry and Forensic Sciences, School of Chemistry and Biochemical Science, C. K. Tedam University of Technology and Applied Sciences, P. O. Box 24, Navrongo, Ghana
- Institute of Neuroscience, Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052,China
| |
Collapse
|
2
|
Zhang S, Zhang S, Fan Y, Zhang X, Chen J, Jin C, Chen S, Wang L, Zhang Q, Chen Y. Total Synthesis of the Proposed Structure of Neaumycin B. Angew Chem Int Ed Engl 2023; 62:e202313186. [PMID: 37889502 DOI: 10.1002/anie.202313186] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 10/28/2023]
Abstract
The total synthesis of the proposed structure of anti-glioblastoma natural product neaumycin B was achieved in 22 steps (longest linear sequence). The synthesis features HCl-mediated [6,6]-spiroketalization, a combination of Krische iridium-catalyzed crotylation, Marshall palladium-catalyzed propargylation, Fürstner nickel-catalyzed regio- and enantioselective vicinal monoprotected diol formation, Brown crotylation and asymmetric halide-aldehyde cycloaddition, so as to establish the challenging contiguous stereocenters.
Collapse
Affiliation(s)
- Sen Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University 38 Tongyan Road, Tianjin 300353 (P. R. China)
| | - Songming Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Frontiers Science Center for New Organic Matter, Nankai University, 94 Weijin Road, Tianjin, 300071, P. R. China
| | - Yunlong Fan
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University 38 Tongyan Road, Tianjin 300353 (P. R. China)
| | - Xuhai Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University 38 Tongyan Road, Tianjin 300353 (P. R. China)
| | - Jing Chen
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University 38 Tongyan Road, Tianjin 300353 (P. R. China)
| | - Chaofan Jin
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University 38 Tongyan Road, Tianjin 300353 (P. R. China)
| | - Sisi Chen
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Frontiers Science Center for New Organic Matter, Nankai University, 94 Weijin Road, Tianjin, 300071, P. R. China
| | - Liang Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Frontiers Science Center for New Organic Matter, Nankai University, 94 Weijin Road, Tianjin, 300071, P. R. China
| | - Quan Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University 38 Tongyan Road, Tianjin 300353 (P. R. China)
| | - Yue Chen
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Frontiers Science Center for New Organic Matter, Nankai University, 94 Weijin Road, Tianjin, 300071, P. R. China
| |
Collapse
|
3
|
López C, Schleussner N, Bernhart SH, Kleinheinz K, Sungalee S, Sczakiel HL, Kretzmer H, Toprak UH, Glaser S, Wagener R, Ammerpohl O, Bens S, Giefing M, González Sánchez JC, Apic G, Hübschmann D, Janz M, Kreuz M, Mottok A, Müller JM, Seufert J, Hoffmann S, Korbel JO, Russell RB, Schüle R, Trümper L, Klapper W, Radlwimmer B, Lichter P, Küppers R, Schlesner M, Mathas S, Siebert R. Focal structural variants revealed by whole genome sequencing disrupt the histone demethylase KDM4C in B-cell lymphomas. Haematologica 2022; 108:543-554. [PMID: 35522148 PMCID: PMC9890021 DOI: 10.3324/haematol.2021.280005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Indexed: 02/03/2023] Open
Abstract
Histone methylation-modifiers, such as EZH2 and KMT2D, are recurrently altered in B-cell lymphomas. To comprehensively describe the landscape of alterations affecting genes encoding histone methylation-modifiers in lymphomagenesis we investigated whole genome and transcriptome data of 186 mature B-cell lymphomas sequenced in the ICGC MMML-Seq project. Besides confirming common alterations of KMT2D (47% of cases), EZH2 (17%), SETD1B (5%), PRDM9 (4%), KMT2C (4%), and SETD2 (4%), also identified by prior exome or RNA-sequencing studies, we here found recurrent alterations to KDM4C in chromosome 9p24, encoding a histone demethylase. Focal structural variation was the main mechanism of KDM4C alterations, and was independent from 9p24 amplification. We also identified KDM4C alterations in lymphoma cell lines including a focal homozygous deletion in a classical Hodgkin lymphoma cell line. By integrating RNA-sequencing and genome sequencing data we predict that KDM4C structural variants result in loss-offunction. By functional reconstitution studies in cell lines, we provide evidence that KDM4C can act as a tumor suppressor. Thus, we show that identification of structural variants in whole genome sequencing data adds to the comprehensive description of the mutational landscape of lymphomas and, moreover, establish KDM4C as a putative tumor suppressive gene recurrently altered in subsets of B-cell derived lymphomas.
Collapse
Affiliation(s)
- Cristina López
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany,Institute of Human Genetics, Christian-Albrechts-University, Kiel, Germany,*CL and NS contributed equally as co-first authors
| | - Nikolai Schleussner
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany,Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany, and Experimental and Clinical Research Center, a joint cooperation between the MDC and the Charité, Berlin, Germany,*CL and NS contributed equally as co-first authors
| | - Stephan H. Bernhart
- Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany,Bioinformatics Group, Department of Computer, University of Leipzig, Leipzig, Germany,Transcriptome Bioinformatics, LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Kortine Kleinheinz
- Department for Bioinformatics and Functional Genomics, Institute of Pharmacy and Molecular Biotechnology and Bioquant, University of Heidelberg, Heidelberg, Germany
| | | | - Henrike L. Sczakiel
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany,Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany, and Experimental and Clinical Research Center, a joint cooperation between the MDC and the Charité, Berlin, Germany
| | - Helene Kretzmer
- Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany,Bioinformatics Group, Department of Computer, University of Leipzig, Leipzig, Germany,Transcriptome Bioinformatics, LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany,Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Umut H. Toprak
- Bioinformatics and Omics Data Analytics (B240), German Cancer Research Center (DKFZ), Heidelberg, Germany,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany,Hopp-Children’s Cancer Center at the NCT Heidelberg (KiTZ), Division of Neuroblastoma Genomics (B087), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Selina Glaser
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Rabea Wagener
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany,Institute of Human Genetics, Christian-Albrechts-University, Kiel, Germany
| | - Ole Ammerpohl
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany,Institute of Human Genetics, Christian-Albrechts-University, Kiel, Germany
| | - Susanne Bens
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany,Institute of Human Genetics, Christian-Albrechts-University, Kiel, Germany
| | - Maciej Giefing
- Institute of Human Genetics, Christian-Albrechts-University, Kiel, Germany,Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | | | - Gordana Apic
- BioQuant and Biochemie Zentrum Heidelberg (BZH), Heidelberg University, Heidelberg, Germany
| | - Daniel Hübschmann
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany,Department for Bioinformatics and Functional Genomics, Institute of Pharmacy and Molecular Biotechnology and Bioquant, University of Heidelberg, Heidelberg, Germany,German Cancer Consortium (DKTK), Heidelberg, Germany,Heidelberg Institute of Stem Cell Technology and Experimental Medicine (HI-STEM), Heidelberg, Germany
| | - Martin Janz
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany,Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany, and Experimental and Clinical Research Center, a joint cooperation between the MDC and the Charité, Berlin, Germany
| | - Markus Kreuz
- Institute for Medical Informatics Statistics and Epidemiology, Leipzig, Germany
| | - Anja Mottok
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Judith M. Müller
- Klinik fur Urologie und Zentrale Klinische Forschung, Klinikum der Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Julian Seufert
- Bioinformatics and Omics Data Analytics (B240), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Steve Hoffmann
- Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany,Bioinformatics Group, Department of Computer, University of Leipzig, Leipzig, Germany,Transcriptome Bioinformatics, LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany,Leibniz Institute on Ageing-Fritz Lipmann Institute (FLI), Computational Biology, Jena, Germany
| | - Jan O. Korbel
- EMBL Heidelberg, Genome Biology Unit, Heidelberg,, Germany
| | - Robert B. Russell
- BioQuant and Biochemie Zentrum Heidelberg (BZH), Heidelberg University, Heidelberg, Germany
| | - Roland Schüle
- Klinik fur Urologie und Zentrale Klinische Forschung, Klinikum der Albert-Ludwigs-Universität Freiburg, Freiburg, Germany,BIOSS Centre of Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Lorenz Trümper
- Department of Hematology and Oncology, Georg-August-University of Göttingen, Göttingen, Germany
| | - Wolfram Klapper
- Hematopathology Section, Christian-Albrechts-University, Kiel, Germany
| | - Bernhard Radlwimmer
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Lichter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ralf Küppers
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Essen, Germany, and German Cancer Consortium (DKTK)
| | - Matthias Schlesner
- Bioinformatics and Omics Data Analytics (B240), German Cancer Research Center (DKFZ), Heidelberg, Germany,Biomedical Informatics, Data Mining and Data Analytics, Augsburg University, Augsburg, Germany
| | - Stephan Mathas
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany,Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany, and Experimental and Clinical Research Center, a joint cooperation between the MDC and the Charité, Berlin, Germany,SM and RS contributed equally as co-senior authors
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany,Institute of Human Genetics, Christian-Albrechts-University, Kiel, Germany,SM and RS contributed equally as co-senior authors
| |
Collapse
|
4
|
Wang T, Zhang R, Liu Y, Fang Z, Zhang H, Fan Y, Yang S, Xiang R. Discovery of a new class of JMJD6 inhibitors and structure-activity relationship study. Bioorg Med Chem Lett 2021; 44:128109. [PMID: 33991627 DOI: 10.1016/j.bmcl.2021.128109] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/10/2021] [Indexed: 02/05/2023]
Abstract
JmjC domain-containing protein 6 (JMJD6) has been thought as a potential target for various diseases particularly cancer. However, few selective JMJD6 inhibitors have been reported. In this investigation, molecular docking and biological activity evaluation were performed to retrieve new JMJD6 inhibitors, which led to the identification of a hit compound, J2. Further structural optimization and structure-activity relationship (SAR) analysis towards J2 were carried out, which gave a new potent JMJD6 inhibitor, 7p. This compound showed an IC50 value of 0.681 μM against JMJD6, but displayed no activity against other tested JmjC domain-containing protein family members, indicating good selectivity (>100 fold). Collectively, this investigation offers a selective JMJD6 inhibitor, which could be taken as a lead compound for subsequent drug discovery targeting JMJD6.
Collapse
Affiliation(s)
- Tianqi Wang
- Department of Medicinal Chemistry, School of Medicine, Nankai University, Tianjin 300071, PR China
| | - Rong Zhang
- West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yang Liu
- West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Zhen Fang
- Department of Medicinal Chemistry, School of Medicine, Nankai University, Tianjin 300071, PR China
| | - Hailin Zhang
- West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yan Fan
- Department of Medicinal Chemistry, School of Medicine, Nankai University, Tianjin 300071, PR China
| | - Shengyong Yang
- West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Rong Xiang
- Department of Medicinal Chemistry, School of Medicine, Nankai University, Tianjin 300071, PR China.
| |
Collapse
|
5
|
Borgenvik A, Čančer M, Hutter S, Swartling FJ. Targeting MYCN in Molecularly Defined Malignant Brain Tumors. Front Oncol 2021; 10:626751. [PMID: 33585252 PMCID: PMC7877538 DOI: 10.3389/fonc.2020.626751] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/09/2020] [Indexed: 12/21/2022] Open
Abstract
Misregulation of MYC genes, causing MYC overexpression or protein stabilization, is frequently found in malignant brain tumors highlighting their important roles as oncogenes. Brain tumors in children are the most lethal of all pediatric malignancies and the most common malignant primary adult brain tumor, glioblastoma, is still practically incurable. MYCN is one of three MYC family members and is crucial for normal brain development. It is associated with poor prognosis in many malignant pediatric brain tumor types and is focally amplified in specific adult brain tumors. Targeting MYCN has proved to be challenging due to its undruggable nature as a transcription factor and for its importance in regulating developmental programs also in healthy cells. In this review, we will discuss efforts made to circumvent the difficulty of targeting MYCN specifically by using direct or indirect measures to treat MYCN-driven brain tumors. We will further consider the mechanism of action of these measures and suggest which molecularly defined brain tumor patients that might benefit from MYCN-directed precision therapies.
Collapse
Affiliation(s)
- Anna Borgenvik
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Matko Čančer
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Sonja Hutter
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Fredrik J Swartling
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
6
|
Yang J, Chen S, Yang Y, Ma X, Shao B, Yang S, Wei Y, Wei X. Jumonji domain-containing protein 6 protein and its role in cancer. Cell Prolif 2020; 53:e12747. [PMID: 31961032 PMCID: PMC7046477 DOI: 10.1111/cpr.12747] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 02/05/2023] Open
Abstract
The jumonji domain‐containing protein 6 (JMJD6) is a Fe(II)‐ and 2‐oxoglutarate (2OG)‐dependent oxygenase that catalyses lysine hydroxylation and arginine demethylation of histone and non‐histone peptides. Recently, the intrinsic tyrosine kinase activity of JMJD6 has also been reported. The JMJD6 has been implicated in embryonic development, cellular proliferation and migration, self‐tolerance induction in the thymus, and adipocyte differentiation. Not surprisingly, abnormal expression of JMJD6 may contribute to the development of many diseases, such as neuropathic pain, foot‐and‐mouth disease, gestational diabetes mellitus, hepatitis C and various types of cancer. In the present review, we summarized the structure and functions of JMJD6, with particular emphasis on the role of JMJD6 in cancer progression.
Collapse
Affiliation(s)
- Jing Yang
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Siyuan Chen
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanfei Yang
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xuelei Ma
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bin Shao
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Shengyong Yang
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuquan Wei
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiawei Wei
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Li D, Li LF, Zhang ZF, Shi WB, Pan L, Liu Y. Two new Bi(III) and Ce(III) chelates incorporating 1,10-phenanthroline-2,9-dicarboxylic acid: structure elucidation and anti-lung cancer activity study. INORG NANO-MET CHEM 2020. [DOI: 10.1080/24701556.2019.1709505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Di Li
- Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, China
| | - Li-Fei Li
- Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, China
- Department of Orthopedics, Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, China
| | - Zhi-Fang Zhang
- Tong Liao City Hospital, Tongliao, Inner Mongolia, China
| | - Wen-Bo Shi
- Department of Orthopedics, People’s Hospital of Changshan, Quzhou, Zhejiang, China
| | - Lei Pan
- Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, China
- Department of Orthopedics, Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, China
| | - Yang Liu
- Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, China
- Department of Orthopedics, Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, China
| |
Collapse
|
8
|
Zhao C, Gao Y, Guo R, Li H, Yang B. Microarray expression profiles and bioinformatics analysis of mRNAs, lncRNAs, and circRNAs in the secondary temozolomide-resistant glioblastoma. Invest New Drugs 2019; 38:1227-1235. [PMID: 31823158 DOI: 10.1007/s10637-019-00884-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 11/26/2019] [Indexed: 11/30/2022]
Abstract
Temozolomide is a first line anti-tumor drug used for the treatment of patients with Glioblastoma multiforme (GBM). However, the drug resistance to temozolomide limits its clinical application. Therefore, novel strategies to overcome chemoresistance are desperately needed for improved treatment of human GBM. Here, we simultaneously detected, for the first time, the expression profiles of mRNAs, lncRNAs, and circRNAs in three pairs of secondary temozolomide-resistant glioblastoma (STRG) and matched primary glioblastoma tissues by microarrays. Using these data, we discovered a total of 92 mRNA, 299 lncRNAs and 53 circRNAs were altered in human glioma tissue after chemotherapy with temozolomide. The functions of differentially expressed lncRNAs, circRNAs were annotated by analysis of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). The results showed that the highest enriched GO terms of the upregulated lncRNAs were embryonic forelimb morphogenesis (BP), extracellular space (CC), and serine-type endopeptidase activity (MF). Meanwhile, GO:0035360(BP), PRC1 complex (CC), and ubiquitin-protein transferase activity (MF) were the highest enriched GO terms targeted by downregulated lncRNAs. The NF-kappa B signaling pathway were significantly enriched in the STRG. However, circRNAs highest enriched GO term was viral process, chromosome, and protein transporter activity, respectively. KEGG pathway analysis showed that circRNAs in the network were enriched in ErbB signaling pathway. Furthermore, we also predicted the potential role of these differentially expressed ncRNAs and constructed a network of lncRNAs-mRNAs and circRNAs-miRNAs to show their interactions. After a series of bioinformatics analyses, we found that low expression of NONHSAT163779 and high expression of circ_0043949 are closely related to the chemoresistance of STRG. Our findings revealed the alteration of expression patterns of mRNAs, lncRNAs, and circRNAs in the secondary temozolomide-resistant glioblastoma for the first time. NONHSAT163779 and hsa_circ_0043949 might be potential therapeutic targets and prognostic biomarkers for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Chengbin Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Rd, Erqi District, Zhengzhou, 450052, Henan, China
| | - Yuyuan Gao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Rd, Erqi District, Zhengzhou, 450052, Henan, China
| | - Ruiming Guo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Rd, Erqi District, Zhengzhou, 450052, Henan, China
| | - Hongwei Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Rd, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Bo Yang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Rd, Erqi District, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
9
|
Angeli D, Fanciulli M, Pallocca M. Reverse Engineering Cancer: Inferring Transcriptional Gene Signatures from Copy Number Aberrations with ICAro. Cancers (Basel) 2019; 11:E256. [PMID: 30813319 PMCID: PMC6406408 DOI: 10.3390/cancers11020256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 02/07/2019] [Accepted: 02/13/2019] [Indexed: 12/12/2022] Open
Abstract
The characterization of a gene product function is a process that involves multiple laboratory techniques in order to silence the gene itself and to understand the resulting cellular phenotype via several omics profiling. When it comes to tumor cells, usually the translation process from in vitro characterization results to human validation is a difficult journey. Here, we present a simple algorithm to extract mRNA signatures from cancer datasets, where a particular gene has been deleted at the genomic level, ICAro. The process is implemented as a two-step workflow. The first one employs several filters in order to select the two patient subsets: the inactivated one, where the target gene is deleted, and the control one, where large genomic rearrangements should be absent. The second step performs a signature extraction via a Differential Expression analysis and a complementary Random Forest approach to provide an additional gene ranking in terms of information loss. We benchmarked the system robustness on a panel of genes frequently deleted in cancers, where we validated the downregulation of target genes and found a correlation with signatures extracted with the L1000 tool, outperforming random sampling for two out of six L1000 classes. Furthermore, we present a use case correlation with a published transcriptomic experiment. In conclusion, deciphering the complex interactions of the tumor environment is a challenge that requires the integration of several experimental techniques in order to create reproducible results. We implemented a tool which could be of use when trying to find mRNA signatures related to a gene loss event to better understand its function or for a gene-loss associated biomarker research.
Collapse
Affiliation(s)
- Davide Angeli
- Department of Paediatric Haematology, IRCCS Ospedale Pediatrico Bambino Gesù, 00146 Rome, Italy.
| | - Maurizio Fanciulli
- SAFU Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy.
| | - Matteo Pallocca
- SAFU Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy.
| |
Collapse
|
10
|
Oksdath M, Perrin SL, Bardy C, Hilder EF, DeForest CA, Arrua RD, Gomez GA. Review: Synthetic scaffolds to control the biochemical, mechanical, and geometrical environment of stem cell-derived brain organoids. APL Bioeng 2018; 2:041501. [PMID: 31069322 PMCID: PMC6481728 DOI: 10.1063/1.5045124] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 10/31/2018] [Indexed: 01/16/2023] Open
Abstract
Stem cell-derived brain organoids provide a powerful platform for systematic studies of tissue functional architecture and the development of personalized therapies. Here, we review key advances at the interface of soft matter and stem cell biology on synthetic alternatives to extracellular matrices. We emphasize recent biomaterial-based strategies that have been proven advantageous towards optimizing organoid growth and controlling the geometrical, biomechanical, and biochemical properties of the organoid's three-dimensional environment. We highlight systems that have the potential to increase the translational value of region-specific brain organoid models suitable for different types of manipulations and high-throughput applications.
Collapse
Affiliation(s)
- Mariana Oksdath
- Centre for Cancer Biology, South Australia Pathology and University of South Australia, Adelaide 5001, Australia
| | - Sally L. Perrin
- Centre for Cancer Biology, South Australia Pathology and University of South Australia, Adelaide 5001, Australia
| | | | - Emily F. Hilder
- Future Industries Institute, University of South Australia, Mawson Lakes 5095, Australia
| | - Cole A. DeForest
- Department of Chemical Engineering and Department of Bioengineering, University of Washington, Seattle, Washington 98195-1750, USA
| | - R. Dario Arrua
- Future Industries Institute, University of South Australia, Mawson Lakes 5095, Australia
| | - Guillermo A. Gomez
- Centre for Cancer Biology, South Australia Pathology and University of South Australia, Adelaide 5001, Australia
| |
Collapse
|
11
|
Li D, Chi G, Chen Z, Jin X. MicroRNA-1225-5p behaves as a tumor suppressor in human glioblastoma via targeting of IRS1. Onco Targets Ther 2018; 11:6339-6350. [PMID: 30319274 PMCID: PMC6167988 DOI: 10.2147/ott.s178001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background MicroRNAs (miRNAs) play an important role in cancer initiation, progression, and metastasis by directly regulating their target genes. Materials and methods In this study, we observed that the miR-1225-5p expression level in glioblastoma tissues was significantly lower as compared with that in normal brain tissues, and its low expression was significantly associated with histopathological grade and poor patient prognosis. Results Through establishing a miR-1225-5p overexpression glioblastoma cell line, we found that ectopic overexpression of miR-1225-5p inhibited the proliferation, migration, and invasion of glioblastoma cells in vitro. Moreover, the growth of a glioblastoma xenograft tumor was attenuated by overexpression of miR-1225-5p. Further integrative studies suggested that the insulin receptor substrate 1 (IRS1) was a direct functional target of miR-1225-5p in glioblastoma, and the mRNA and protein levels of IRS1 in six human glioblastoma cell lines (A172, SW1783, U87, LN-229, SW1088, and T98G) were significantly higher as compared with normal human astrocytes. Conclusion These results suggest that miR-1225-5p may be a novel candidate for glioblastoma therapy.
Collapse
Affiliation(s)
- Dongyuan Li
- First Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, People's Republic of China,
| | - Guonan Chi
- First Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, People's Republic of China,
| | - Zhuo Chen
- First Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, People's Republic of China,
| | - Xingyi Jin
- First Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, People's Republic of China,
| |
Collapse
|
12
|
Sun L, Yang S, Chi G, Jin X. Hsp90 inhibitor NMS-E973 exerts the anticancer effect against glioblastoma via induction of PUMA-mediated apoptosis. Onco Targets Ther 2018; 11:1583-1593. [PMID: 29593424 PMCID: PMC5865573 DOI: 10.2147/ott.s160813] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Glioblastoma is one of the most aggressive and common malignancies of the central nervous system in humans. Owing to the correlation of high Hsp90 expression with prognosis and clinical pathology features of diverse types of cancer, targeting Hsp90 with small-molecule inhibitors has become a promising anticancer strategy. PURPOSE In this study, we aimed to explore the possibility of anticancer effect of NMS-E973 in giloblastoma and elucidate the mechanism. METHODS Cell based MTT assay and colony formation assay were used to detect cell viability. Apoptosis was analyzed by nuclear staining with Hoechst 33258 and Annexin V/propidium iodide staining followed by flow cytometry. Western-blot and RT-PCR were used to detect gene expression. Xenograft assay was used to explore the anticancer effect of NMS-E973 in vivo. RESULTS We found that NMS-E973 induces apoptosis and inhibits cell growth in glioblastoma cells in cell culture and xenograft models. As a proapoptotic Bcl-2 member, PUMA was induced by NMS-E973 in a p53-dependent manner in glioblastoma in cell culture, thereby inducing apoptosis in glioblastoma cells. Furthermore, PUMA was induced by NMS-E973 treatment in xenograft tumors, and deficiency in PUMA significantly suppressed the antitumor effects of NMS-E973. CONCLUSION Our study suggests that PUMA-mediated apoptosis is important for the therapeutic responses of NMS-E973. Induction of PUMA might be a potential biomarker for predicting NMS-E973 responses.
Collapse
Affiliation(s)
- Libo Sun
- First Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changhun, Jilin, People’s Republic of China
| | - Shoujun Yang
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changhun, Jilin, People’s Republic of China
| | - Guonan Chi
- First Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changhun, Jilin, People’s Republic of China
| | - Xingyi Jin
- First Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changhun, Jilin, People’s Republic of China
| |
Collapse
|