1
|
Budhiraja S, McManus G, Baisiwala S, Perrault EN, Cho S, Saathoff M, Chen L, Park CH, Kazi HA, Dmello C, Lin P, James CD, Sonabend AM, Heiland DH, Ahmed AU. ARF4-mediated retrograde trafficking as a driver of chemoresistance in glioblastoma. Neuro Oncol 2024; 26:1421-1437. [PMID: 38506351 PMCID: PMC11300013 DOI: 10.1093/neuonc/noae059] [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: 10/17/2023] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Cellular functions hinge on the meticulous orchestration of protein transport, both spatially and temporally. Central to this process is retrograde trafficking, responsible for targeting proteins to the nucleus. Despite its link to many diseases, the implications of retrograde trafficking in glioblastoma (GBM) are still unclear. METHODS To identify genetic drivers of TMZ resistance, we conducted comprehensive CRISPR-knockout screening, revealing ADP-ribosylation factor 4 (ARF4), a regulator of retrograde trafficking, as a major contributor. RESULTS Suppressing ARF4 significantly enhanced TMZ sensitivity in GBM patient-derived xenograft (PDX) models, leading to improved survival rates (P < .01) in both primary and recurrent lines. We also observed that TMZ exposure stimulates ARF4-mediated retrograde trafficking. Proteomics analysis of GBM cells with varying levels of ARF4 unveiled the influence of this pathway on EGFR signaling, with increased nuclear trafficking of EGFR observed in cells with ARF4 overexpression and TMZ treatment. Additionally, spatially resolved RNA-sequencing of GBM patient tissues revealed substantial correlations between ARF4 and crucial nuclear EGFR (nEGFR) downstream targets, such as MYC, STAT1, and DNA-PK. Decreased activity of DNA-PK, a DNA repair protein downstream of nEGFR signaling that contributes to TMZ resistance, was observed in cells with suppressed ARF4 levels. Notably, treatment with DNA-PK inhibitor, KU-57788, in mice with a recurrent PDX line resulted in prolonged survival (P < .01), highlighting the promising therapeutic implications of targeting proteins reliant on ARF4-mediated retrograde trafficking. CONCLUSIONS Our findings demonstrate that ARF4-mediated retrograde trafficking contributes to the development of TMZ resistance, cementing this pathway as a viable strategy to overcome chemoresistance in GBM.
Collapse
Affiliation(s)
- Shreya Budhiraja
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Graysen McManus
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - Ella N Perrault
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Sia Cho
- Department of Neurobiology, Northwestern University, Evanston, Illinois, USA
| | - Miranda Saathoff
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Li Chen
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Cheol H Park
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Hasaan A Kazi
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Crismita Dmello
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Peiyu Lin
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - C David James
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Adam M Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Dieter H Heiland
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Microenvironment and Immunology Research Laboratory, Medical Center - University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Freiburg, Germany
| | - Atique U Ahmed
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
2
|
He L, Azizad D, Bhat K, Ioannidis A, Hoffmann CJ, Arambula E, Bhaduri A, Kornblum HI, Pajonk F. Radiation-Induced Cellular Plasticity: A Strategy for Combatting Glioblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.593985. [PMID: 38798647 PMCID: PMC11118449 DOI: 10.1101/2024.05.13.593985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Glioblastoma is the deadliest brain cancer in adults and almost all patients succumb to the tumor. While surgery followed by chemo-radiotherapy significantly delays disease progression, these treatments do not lead to long-term tumor control and targeted therapies or biologics have so far failed to further improve survival. Utilizing a transient radiation-induced state of multipotency we used the adenylcyclase activator forskolin to alter the cellular fate of glioma cells in response to radiation. The combined treatment induced the expression of neuronal markers in glioma cells, reduced proliferation and led to a distinct gene expression profile. scRNAseq revealed that the combined treatment forced glioma cells into a microglia- and neuron-like phenotypes. In vivo this treatment led to a loss of glioma stem cells and prolonged median survival in mouse models of glioblastoma. Collectively, our data suggest that revisiting a differentiation therapy with forskolin in combination with radiation could lead to clinical benefit.
Collapse
Affiliation(s)
- Ling He
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA
- Jonsson Comprehensive Cancer Center at UCLA
| | | | - Kruttika Bhat
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA
| | - Angeliki Ioannidis
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA
| | - Carter J. Hoffmann
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA
| | - Evelyn Arambula
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA
| | - Aparna Bhaduri
- Jonsson Comprehensive Cancer Center at UCLA
- Department of Biological Chemistry at UCLA
| | - Harley I. Kornblum
- Jonsson Comprehensive Cancer Center at UCLA
- NPI-Semel Institute for Neuroscience & Human Behavior at UCLA
| | - Frank Pajonk
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA
- Jonsson Comprehensive Cancer Center at UCLA
- Department of Neurosurgery, David Geffen School of Medicine at UCLA
| |
Collapse
|
3
|
Abdolahi M, Ghaedi Talkhounche P, Derakhshan Nazari MH, Hosseininia HS, Khoshdel-Rad N, Ebrahimi Sadrabadi A. Functional Enrichment Analysis of Tumor Microenvironment-Driven Molecular Alterations That Facilitate Epithelial-to-Mesenchymal Transition and Distant Metastasis. Bioinform Biol Insights 2024; 18:11779322241227722. [PMID: 38318286 PMCID: PMC10840405 DOI: 10.1177/11779322241227722] [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: 01/06/2023] [Accepted: 01/04/2024] [Indexed: 02/07/2024] Open
Abstract
Nowadays, hepatocellular carcinoma (HCC) is the second leading cause of cancer deaths, and identifying the effective factors in causing this disease can play an important role in its prevention and treatment. Tumors provide effective agents for invasion and metastasis to other organs by establishing appropriate communication between cancer cells and the microenvironment. Epithelial-to-mesenchymal transition (EMT) can be mentioned as one of the effective phenomena in tumor invasion and metastasis. Several factors are involved in inducing this phenomenon in the tumor microenvironment, which helps the tumor survive and migrate to other places. It can be effective to identify these factors in the use of appropriate treatment strategies and greater patient survival. This study investigated the molecular differences between tumor border cells and tumor core cells or internal tumor cells in HCC for specific EMT genes. Expression of NOTCH1, ID1, and LST1 genes showed a significant increase at the HCC tumor border. Targeting these genes can be considered as a useful therapeutic strategy to prevent distant metastasis in HCC patients.
Collapse
Affiliation(s)
- Mahnaz Abdolahi
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University, Tehran, Iran
| | - Parnian Ghaedi Talkhounche
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Mohammad Hossein Derakhshan Nazari
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Haniyeh Sadat Hosseininia
- Department of Cellular and Molecular Biology, Faculty of Advanced Medical Science, Islamic Azad University of Medical Sciences, Tehran, Iran
- Cytotech & Bioinformatics Research Group, Bioinformatics Department, Tehran, Iran
| | - Niloofar Khoshdel-Rad
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Amin Ebrahimi Sadrabadi
- Cytotech & Bioinformatics Research Group, Bioinformatics Department, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACER, Tehran, Iran
| |
Collapse
|
4
|
Golán-Cancela I, Caja L. The TGF-β Family in Glioblastoma. Int J Mol Sci 2024; 25:1067. [PMID: 38256140 PMCID: PMC10816220 DOI: 10.3390/ijms25021067] [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: 11/14/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Members of the transforming growth factor β (TGF-β) family have been implicated in the biology of several cancers. In this review, we focus on the role of TGFβ and bone morphogenetic protein (BMP) signaling in glioblastoma. Glioblastoma (GBM) is the most common malignant brain tumor in adults; it presents at a median age of 64 years, but can occur at any age, including childhood. Unfortunately, there is no cure, and even patients undergoing current treatments (surgical resection, radiotherapy, and chemotherapy) have a median survival of 15 months. There is a great need to identify new therapeutic targets to improve the treatment of GBM patients. TGF-βs signaling promotes tumorigenesis in glioblastoma, while BMPs suppress tumorigenic potential by inducing tumor cell differentiation. In this review, we discuss the actions of TGF-βs and BMPs on cancer cells as well as in the tumor microenvironment, and their use in potential therapeutic intervention.
Collapse
Affiliation(s)
| | - Laia Caja
- Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, SE-75123 Uppsala, Sweden;
| |
Collapse
|
5
|
Li ZX, Sun MC, Fang K, Zhao ZY, Leng ZY, Zhang ZH, Xu AP, Chu Y, Zhang L, Lian J, Chen T, Xu MD. Transcription factor 3 promotes migration and invasion potential and maintains cancer stemness by activating ID1 expression in esophageal squamous cell carcinoma. Cancer Biol Ther 2023; 24:2246206. [PMID: 37607071 PMCID: PMC10443991 DOI: 10.1080/15384047.2023.2246206] [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: 03/14/2022] [Revised: 10/09/2022] [Accepted: 06/06/2023] [Indexed: 08/24/2023] Open
Abstract
Transcription factor 3 (TCF3) is a member of the basic Helix - Loop - Helix (bHLH) transcription factor (TF) family and is encoded by the TCF3 gene (also known as E2A). It has been shown that TCF3 functions as a key transcription factor in the pathogenesis of several human cancers and plays an important role in stem cell maintenance and carcinogenesis. However, the effect of TCF3 in the progression of esophageal squamous cell carcinoma (ESCC) is poorly known. In our study, TCF3 was found to express highly and correlated with cancer stage and prognosis. TCF3 was shown to promote ESCC invasion, migration, and drug resistance both from the results of in vivo and in vitro assays. Moreover, further studies suggested that TCF3 played these roles through transcriptionally regulating Inhibitor of DNA binding 1(ID1). Notably, we also found that TCF3 or ID1 was associated with ESCC stemness. Furthermore, TCF3 was correlated with the expression of cancer stemness markers CD44 and CD133. Therefore, maintaining cancer stemness might be the underlying mechanism that TCF3 transcriptionally regulated ID1 and further promoted ESCC progression and drug resistance.
Collapse
Affiliation(s)
- Zhao-Xing Li
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ming-Chuang Sun
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Kang Fang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zi-Ying Zhao
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhu-Yun Leng
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ze-Hua Zhang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ai-Ping Xu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuan Chu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Li Zhang
- Department of Pathology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jingjing Lian
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tao Chen
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Mei-Dong Xu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
6
|
Song P, Yue Q, Chen X, Fu Q, Zhang P, Zhou R. Identification of ID1 and miR-150 interaction and effects on proliferation and apoptosis in ovine granulosa cells. Theriogenology 2023; 212:1-8. [PMID: 37672890 DOI: 10.1016/j.theriogenology.2023.08.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 08/19/2023] [Accepted: 08/31/2023] [Indexed: 09/08/2023]
Abstract
Granulosa cells (GCs) proliferation and apoptosis play a significantly role in follicular development and atresia. ID1 and miR-150 are involved in cell apoptosis and follicular atresia, but the interaction and function of ID1 and miR-150 in GCs are unclear. This study focuses on ID1 and miR-150 in terms of the interaction and effects on proliferation and apoptosis in ovine granulosa cells. Our findings revealed that ID1 decreased the promoter activity and expression level of oar-miR-150. However, the expression of ID1 was downregulated by miR-150, and ID1 was identified as a target gene of oar-miR-150. miR-150 mimic inhibited proliferation and upregulated apoptosis rate in ovine GCs, while the results of miR-150 inhibitor were opposite. Overexpression of ID1 significantly inhibited ovine GCs proliferation and cell cycle-related genes (CDK1, CDK2, CDK4, CCND2, CDC20, and PCNA) expression, whereas knockdown of ID1 promoted cell proliferation and those genes expression. Overexpression of ID1 significantly downregulated mitochondrial membrane potential and Bcl-2 expression in ovine GCs, and upregulated the expression of pro-apoptosis genes Bax, Caspase-3, and Caspase-9, whereas the results of ID1 knockdown were reversed. Collectively, these findings indicate the interaction and the vital role of ID1 and miR-150 on proliferation and apoptosis in ovine granulosa cells, which may suggest a novel target for ovine follicular development and atresia.
Collapse
Affiliation(s)
- Pengyan Song
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, Hebei Province, 071001, China
| | - Qiaoxian Yue
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, Hebei Province, 071001, China
| | - Xiaoyong Chen
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, Hebei Province, 071001, China
| | - Qiang Fu
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, Hebei Province, 071001, China
| | - Peiying Zhang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, Hebei Province, 071001, China
| | - Rongyan Zhou
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, Hebei Province, 071001, China.
| |
Collapse
|
7
|
Nasrolahi A, Azizidoost S, Radoszkiewicz K, Najafi S, Ghaedrahmati F, Anbiyaee O, Khoshnam SE, Farzaneh M, Uddin S. Signaling pathways governing glioma cancer stem cells behavior. Cell Signal 2023; 101:110493. [PMID: 36228964 DOI: 10.1016/j.cellsig.2022.110493] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/30/2022]
Abstract
Glioma is the most common malignant brain tumor that develops in the glial tissue. Several studies have identified that glioma cancer stem cells (GCSCs) play important roles in tumor-initiating features in malignant gliomas. GCSCs are a small population in the brain that presents an essential role in the metastasis of glioma cells to other organs. These cells can self-renew and differentiate, which are thought to be involved in the pathogenesis of glioma. Therefore, targeting GCSCs might be a novel strategy for the treatment of glioma. Accumulating evidence revealed that several signaling pathways, including Notch, TGF-β, Wnt, STAT3, AKT, and EGFR mediated GCSC growth, proliferation, migration, and invasion. Besides, non-coding RNAs (ncRNAs), including miRNAs, circular RNAs, and long ncRNAs have been found to play pivotal roles in the regulation of GCSC pathogenesis and drug resistance. Therefore, targeting these pathways could open a new avenue for glioma management. In this review, we summarized critical signaling pathways involved in the stimulation or prevention of GCSCs tumorigenesis and invasiveness.
Collapse
Affiliation(s)
- Ava Nasrolahi
- Infectious Ophthalmologic Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shirin Azizidoost
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Klaudia Radoszkiewicz
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Poland
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Omid Anbiyaee
- Cardiovascular Research Center, Nemazi Hospital, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Shahab Uddin
- Translational Research Institute and Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.
| |
Collapse
|
8
|
Hersh AM, Gaitsch H, Alomari S, Lubelski D, Tyler BM. Molecular Pathways and Genomic Landscape of Glioblastoma Stem Cells: Opportunities for Targeted Therapy. Cancers (Basel) 2022; 14:3743. [PMID: 35954407 PMCID: PMC9367289 DOI: 10.3390/cancers14153743] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 02/01/2023] Open
Abstract
Glioblastoma (GBM) is an aggressive tumor of the central nervous system categorized by the World Health Organization as a Grade 4 astrocytoma. Despite treatment with surgical resection, adjuvant chemotherapy, and radiation therapy, outcomes remain poor, with a median survival of only 14-16 months. Although tumor regression is often observed initially after treatment, long-term recurrence or progression invariably occurs. Tumor growth, invasion, and recurrence is mediated by a unique population of glioblastoma stem cells (GSCs). Their high mutation rate and dysregulated transcriptional landscape augment their resistance to conventional chemotherapy and radiation therapy, explaining the poor outcomes observed in patients. Consequently, GSCs have emerged as targets of interest in new treatment paradigms. Here, we review the unique properties of GSCs, including their interactions with the hypoxic microenvironment that drives their proliferation. We discuss vital signaling pathways in GSCs that mediate stemness, self-renewal, proliferation, and invasion, including the Notch, epidermal growth factor receptor, phosphatidylinositol 3-kinase/Akt, sonic hedgehog, transforming growth factor beta, Wnt, signal transducer and activator of transcription 3, and inhibitors of differentiation pathways. We also review epigenomic changes in GSCs that influence their transcriptional state, including DNA methylation, histone methylation and acetylation, and miRNA expression. The constituent molecular components of the signaling pathways and epigenomic regulators represent potential sites for targeted therapy, and representative examples of inhibitory molecules and pharmaceuticals are discussed. Continued investigation into the molecular pathways of GSCs and candidate therapeutics is needed to discover new effective treatments for GBM and improve survival.
Collapse
Affiliation(s)
- Andrew M. Hersh
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.M.H.); (H.G.); (S.A.); (D.L.)
| | - Hallie Gaitsch
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.M.H.); (H.G.); (S.A.); (D.L.)
- NIH Oxford-Cambridge Scholars Program, Wellcome—MRC Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 1TN, UK
| | - Safwan Alomari
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.M.H.); (H.G.); (S.A.); (D.L.)
| | - Daniel Lubelski
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.M.H.); (H.G.); (S.A.); (D.L.)
| | - Betty M. Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.M.H.); (H.G.); (S.A.); (D.L.)
| |
Collapse
|
9
|
Lüke F, Harrer DC, Pantziarka P, Pukrop T, Ghibelli L, Gerner C, Reichle A, Heudobler D. Drug Repurposing by Tumor Tissue Editing. Front Oncol 2022; 12:900985. [PMID: 35814409 PMCID: PMC9270020 DOI: 10.3389/fonc.2022.900985] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
The combinatory use of drugs for systemic cancer therapy commonly aims at the direct elimination of tumor cells through induction of apoptosis. An alternative approach becomes the focus of attention if biological changes in tumor tissues following combinatory administration of regulatorily active drugs are considered as a therapeutic aim, e.g., differentiation, transdifferentiation induction, reconstitution of immunosurveillance, the use of alternative cell death mechanisms. Editing of the tumor tissue establishes new biological 'hallmarks' as a 'pressure point' to attenuate tumor growth. This may be achieved with repurposed, regulatorily active drug combinations, often simultaneously targeting different cell compartments of the tumor tissue. Moreover, tissue editing is paralleled by decisive functional changes in tumor tissues providing novel patterns of target sites for approved drugs. Thus, agents with poor activity in non-edited tissue may reveal new clinically meaningful outcomes. For tissue editing and targeting edited tissue novel requirements concerning drug selection and administration can be summarized according to available clinical and pre-clinical data. Monoactivity is no pre-requisite, but combinatory bio-regulatory activity. The regulatorily active dose may be far below the maximum tolerable dose, and besides inhibitory active drugs stimulatory drug activities may be integrated. Metronomic scheduling often seems to be of advantage. Novel preclinical approaches like functional assays testing drug combinations in tumor tissue are needed to select potential drugs for repurposing. The two-step drug repurposing procedure, namely establishing novel functional systems states in tumor tissues and consecutively providing novel target sites for approved drugs, facilitates the systematic identification of drug activities outside the scope of any original clinical drug approvals.
Collapse
Affiliation(s)
- Florian Lüke
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Dennis Christoph Harrer
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Pan Pantziarka
- The George Pantziarka TP53 Trust, London, United Kingdom
| | - Tobias Pukrop
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), University Hospital Regensburg, Regensburg, Germany
| | - Lina Ghibelli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Albrecht Reichle
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Daniel Heudobler
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
10
|
Kaye J, Mondal A, Foty R, Jia D, Langenfeld J. Bone morphogenetic protein receptor inhibitors suppress the growth of glioblastoma cells. Mol Cell Biochem 2022; 477:1583-1595. [PMID: 35192123 PMCID: PMC8989651 DOI: 10.1007/s11010-022-04383-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 01/31/2022] [Indexed: 01/13/2023]
Abstract
Glioblastomas (GBMs) are aggressive brain tumors that are resistant to chemotherapy and radiation. Bone morphogenetic protein (BMP) ligand BMP4 is being examined as a potential therapeutic for GBMs because it induces differentiation of cancer stem cells (CSCs) to an astrocyte phenotype. ID1 is reported to promote self-renewal and inhibit CSC differentiation. In most cancers, ID1 is transcriptionally upregulated by BMP4 promoting invasion and stemness. This conflicting data bring into question whether BMP signaling is growth suppressive or growth promoting in GBMs. We utilized BMP inhibitors DMH1, JL5, and Ym155 to examine the role of BMP signaling on the growth of GBMs. DMH1 targets BMP type 1 receptors whereas JL5 inhibits both the type 1 and type 2 BMP receptors. Ym155 does not bind the BMP receptors but rather inhibits BMP signaling by inducing the degradation of BMPR2. We show that JL5, DMH1, and Ym155 decreased the expression of ID1 in SD2 and U87 cells. JL5 and Ym155 also decreased the expression of BMPR2 and its downstream target inhibitor of apoptosis protein XIAP. JL5 treatment resulted in significant cell death and suppressed self-renewal to a greater extent than that induced by BMP4 ligand. The lysosome inhibitor chloroquine increases the localization of BMPR2 to the plasma membrane enhancing JL5-induced downregulation of ID1 and cell death in SD2 cells. We show that BMP signaling is growth promoting in GBMs. These studies suggest the need for development of BMP inhibitors and evaluation as potential therapeutic for GBMs.
Collapse
Affiliation(s)
- Joel Kaye
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, The State University of New Jersey, New Brunswick, NJ, 08903, USA
| | - Arindam Mondal
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, The State University of New Jersey, New Brunswick, NJ, 08903, USA
| | - Ramsey Foty
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, The State University of New Jersey, New Brunswick, NJ, 08903, USA
| | - Dongxuan Jia
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, The State University of New Jersey, New Brunswick, NJ, 08903, USA
| | - John Langenfeld
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, The State University of New Jersey, New Brunswick, NJ, 08903, USA.
| |
Collapse
|
11
|
Qiu J, Li Y, Wang B, Sun X, Qian D, Ying Y, Zhou J. The Role and Research Progress of Inhibitor of Differentiation 1 in Atherosclerosis. DNA Cell Biol 2022; 41:71-79. [PMID: 35049366 PMCID: PMC8863915 DOI: 10.1089/dna.2021.0745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/29/2021] [Accepted: 09/29/2021] [Indexed: 12/23/2022] Open
Abstract
Inhibitor of differentiation 1 has a helix-loop-helix (HLH) structure, belongs to a class of molecules known as the HLH trans-acting factor family, and plays an important role in advancing the cell cycle, promoting cell proliferation and inhibiting cell differentiation. Recent studies have confirmed that inhibitor of differentiation 1 plays an important role in the endothelial-mesenchymal transition of vascular endothelial cells, angiogenesis, reendothelialization after injury, and the formation and rupture of atherosclerotic plaques. An in-depth understanding of the role of inhibitor of differentiation 1 in atherosclerosis will provide new ideas and strategies for the treatment of related diseases.
Collapse
Affiliation(s)
- Jun Qiu
- Department of Cardiology, Medicine School of Ningbo University, Ningbo, China
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
- Department of Cardiology, Ningbo Institute of Innovation for Combined Medicine and Engineering (NIIME), Ningbo, China
| | - Youhong Li
- Department of Cardiology, Medicine School of Ningbo University, Ningbo, China
| | - BingYu Wang
- Department of Cardiology, Medicine School of Ningbo University, Ningbo, China
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
- Department of Cardiology, Ningbo Institute of Innovation for Combined Medicine and Engineering (NIIME), Ningbo, China
| | - XinYi Sun
- Department of Cardiology, Medicine School of Ningbo University, Ningbo, China
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
- Department of Cardiology, Ningbo Institute of Innovation for Combined Medicine and Engineering (NIIME), Ningbo, China
| | - Dingding Qian
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Yuchen Ying
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Jianqing Zhou
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| |
Collapse
|
12
|
Tao X, Cheng J, Wang X. Effect of miRNA-200a on radiosensitivity of osteosarcoma cells by targeting Bone morphogenetic protein receptor 2. Bioengineered 2021; 12:12625-12635. [PMID: 34903128 PMCID: PMC8810064 DOI: 10.1080/21655979.2021.2011015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
To study the effect of miR-200a on radiosensitivity of osteosarcoma cells and its mechanism. NC (normal cell) group, mimic-NC group, mimic-miR-200a group, inhibitor-NC group, inhibitor-miR-200a group, si-NC group, si-BMPR2 (Bone morphogenetic protein receptor 2) group, mimic-miR-200a+vector-NC group, and mimic-miR-200a+vector-BMPR2 group were set; the cells of the above groups were irradiated with different radiation intensities (0, 1, 2, 3, and 4 Gy). The expression of miR-200a and BMPR2 mRNA was detected by qRT-PCR; the expression of BMPR2 protein was detected by Western blot; cell viability was detected by MMT (3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide); apoptosis rate was detected by flow cytometry. Cell clone formation experiment was used to detect cell radiosensitivity. Dual-luciferase reporter gene test was used to detect cell fluorescence activity. The expression of BMPR2 was high and the expression of miR-200a was low in osteosarcoma tissues after radiotherapy and in osteosarcoma cells after irradiation. Overexpression of miR-200a and interference with BMPR2 expression inhibits osteosarcoma cell proliferation, promotes apoptosis, and increases cellular radiosensitivity, miR-200a targets expression of BMPR2, and overexpression of BMPR2 reverses the radiosensitizing and apoptotic effects of miR-200a expression on osteosarcoma cells. Overexpression of miR-200a inhibits osteosarcoma cell proliferation, promotes apoptosis, and increases cellular radiosensitivity. The mechanism may be related to the regulation of BMPR2, which may provide new targets and new ideas for osteosarcoma treatment.
Collapse
Affiliation(s)
- Xian Tao
- Department of Orthopedics, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, Jiangsu, China
| | | | - Xinghua Wang
- Department of Orthopedics, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, Jiangsu, China
| |
Collapse
|
13
|
Wang D, Wang D, Huang M, Zheng X, Shen Y, Fu B, Zhao H, Chen X, Peng P, Zhu Q, Zhou Y, Zhang J, Tian Z, Guan W, Wang G, Wei H. Transcriptomic characteristics and impaired immune function of patients who retest positive for SARS-CoV-2 RNA. J Mol Cell Biol 2021; 13:748-759. [PMID: 34687295 PMCID: PMC8574305 DOI: 10.1093/jmcb/mjab067] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 09/02/2021] [Accepted: 09/05/2021] [Indexed: 11/28/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, has become a global public health crisis. Some patients who have recovered from COVID-19 subsequently test positive again for SARS-CoV-2 RNA after discharge from hospital. How such retest-positive (RTP) patients become infected again is not known. In this study, 30 RTP patients, 20 convalescent patients, and 20 healthy controls were enrolled for the analysis of immunological characteristics of their peripheral blood mononuclear cells. We found that absolute numbers of CD4+ T cells, CD8+ T cells, and natural killer cells were not substantially decreased in RTP patients, but the expression of activation markers on these cells was significantly reduced. The percentage of granzyme B-producing T cells was also lower in RTP patients than in convalescent patients. Through transcriptome sequencing, we demonstrated that high expression of inhibitor of differentiation 1 (ID1) and low expression of interferon-induced transmembrane protein 10 (IFITM10) were associated with insufficient activation of immune cells and the occurrence of RTP. These findings provide insight into the impaired immune function associated with COVID-19 and the pathogenesis of RTP, which may contribute to a better understanding of the mechanisms underlying RTP.
Collapse
Affiliation(s)
- Dongyao Wang
- Department of Hematology, The First Affiliated Hospital of the University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.,Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei 230001, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei 230001, China
| | - Dong Wang
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei 230001, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei 230001, China
| | - Min Huang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, HuaZhong University of Science and Technology, Wuhan 430030, China
| | - Xiaohu Zheng
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei 230001, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei 230001, China
| | - Yiqing Shen
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei 230001, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei 230001, China
| | - Binqing Fu
- Department of Hematology, The First Affiliated Hospital of the University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.,Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei 230001, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei 230001, China
| | - Hong Zhao
- Department of Infectious Diseases, Peking University First Hospital, Beijing 100034, China
| | - Xianxiang Chen
- Department of Tuberculosis, Wuhan Pulmonary Hospital, Wuhan 430030, China
| | - Peng Peng
- Department of Tuberculosis, Wuhan Pulmonary Hospital, Wuhan 430030, China
| | - Qi Zhu
- Department of Tuberculosis, Wuhan Pulmonary Hospital, Wuhan 430030, China
| | - Yonggang Zhou
- Department of Hematology, The First Affiliated Hospital of the University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.,Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei 230001, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei 230001, China
| | - Jinghe Zhang
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei 230001, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei 230001, China
| | - Zhigang Tian
- Department of Hematology, The First Affiliated Hospital of the University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.,Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei 230001, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei 230001, China
| | - Wuxiang Guan
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Guiqiang Wang
- Department of Infectious Diseases, Peking University First Hospital, Beijing 100034, China.,Peking University International Hospital, Beijing 100034, China
| | - Haiming Wei
- Department of Hematology, The First Affiliated Hospital of the University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.,Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei 230001, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei 230001, China
| |
Collapse
|
14
|
Chalmers SJ, Murphy SJ, Thompson LL, Hoppman NL, Smadbeck JB, Balcom JR, Harris FR, Frantz RP, Vasmatzis G, E Wylam M. Mate-pair sequencing identifies a cryptic BMPR2 mutation in hereditary pulmonary arterial hypertension. Pulm Circ 2021; 10:2045894020933081. [PMID: 34290857 PMCID: PMC8278463 DOI: 10.1177/2045894020933081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 05/11/2020] [Indexed: 11/29/2022] Open
Abstract
Current guidelines suggest screening all patients with idiopathic pulmonary arterial hypertension for genetic aberrations, particularly mutations in Bone Morphogenic Protein Receptor Type II (BMPR2), the gene most commonly implicated in the pathogenesis of PAH. Herein, we present a novel technique used to identify a pathogenic germline BMPR2 alteration in a 36-year-old female and family members with hereditary pulmonary arterial hypertension who each screened negative by standard cytogenetics and molecular genetics testing.
Collapse
Affiliation(s)
- Sarah J Chalmers
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Stephen J Murphy
- Biomarker Discovery Program, Center for Individualized Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Laura L Thompson
- Department of Laboratory Genetics and Genomics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Nicole L Hoppman
- Department of Laboratory Genetics and Genomics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - James B Smadbeck
- Biomarker Discovery Program, Center for Individualized Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Jessica R Balcom
- Department of Laboratory Genetics and Genomics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Faye R Harris
- Biomarker Discovery Program, Center for Individualized Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Robert P Frantz
- Department of Cardiovascular Disease, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - George Vasmatzis
- Biomarker Discovery Program, Center for Individualized Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Mark E Wylam
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
15
|
Zhou Q, Mei YD, Yang HJ, Tao YL. Inhibitor of DNA-binding family regulates the prognosis of ovarian cancer. Future Oncol 2021; 17:1889-1906. [PMID: 33728938 DOI: 10.2217/fon-2020-1006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: The mechanistic role of inhibitor of DNA binding or differentiation (ID) family in ovarian cancer (OC) has remained unclear. Materials & methods: We used the Oncomine, GEPIA, Kaplan-Meier Plotter, cBioPortal, SurvExpress, PROGgene V2, TIMER, and FunRich to evaluate the prognostic value of IDs in patients with OC. Results: the mRNA transcripts of all IDs were markedly downregulated in OC compared with normal tissue. The prognostic value of IDs was also explored within the subtypes, pathological stages, clinical stages and TP53 mutational status. The group with low-risk IDs showed relatively good overall survival (OS) compared with the high-risk group. Conclusion: ID1/3/4 may be exploited as promising prognostic biomarkers and therapeutic targets in OC patients.
Collapse
Affiliation(s)
- Quan Zhou
- Department of Gynecology & Obstetrics, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, Hubei, 443000, PR China
| | - Ye-Dong Mei
- Department of Obstetrics & Gynecology, The People's Hospital of Wufeng Tujia Autonomous County, Yi Chang, Hubei, 443000, PR China
| | - Huai-Jie Yang
- Department of Gynecology & Obstetrics, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, Hubei, 443000, PR China
| | - Ya-Ling Tao
- Department of Gynecology & Obstetrics, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, Hubei, 443000, PR China
| |
Collapse
|
16
|
Zhou HM, Zhang JG, Zhang X, Li Q. Targeting cancer stem cells for reversing therapy resistance: mechanism, signaling, and prospective agents. Signal Transduct Target Ther 2021; 6:62. [PMID: 33589595 PMCID: PMC7884707 DOI: 10.1038/s41392-020-00430-1] [Citation(s) in RCA: 217] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/26/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs) show a self-renewal capacity and differentiation potential that contribute to tumor progression and therapy resistance. However, the underlying processes are still unclear. Elucidation of the key hallmarks and resistance mechanisms of CSCs may help improve patient outcomes and reduce relapse by altering therapeutic regimens. Here, we reviewed the identification of CSCs, the intrinsic and extrinsic mechanisms of therapy resistance in CSCs, the signaling pathways of CSCs that mediate treatment failure, and potential CSC-targeting agents in various tumors from the clinical perspective. Targeting the mechanisms and pathways described here might contribute to further drug discovery and therapy.
Collapse
Affiliation(s)
- He-Ming Zhou
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of medicine, No.100 Haining Road, 200080, Shanghai, People's Republic of China
| | - Ji-Gang Zhang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of medicine, No.100 Haining Road, 200080, Shanghai, People's Republic of China
| | - Xue Zhang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of medicine, No.100 Haining Road, 200080, Shanghai, People's Republic of China
| | - Qin Li
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of medicine, No.100 Haining Road, 200080, Shanghai, People's Republic of China.
| |
Collapse
|
17
|
Vieira de Castro J, Gonçalves CS, Hormigo A, Costa BM. Exploiting the Complexities of Glioblastoma Stem Cells: Insights for Cancer Initiation and Therapeutic Targeting. Int J Mol Sci 2020; 21:ijms21155278. [PMID: 32722427 PMCID: PMC7432229 DOI: 10.3390/ijms21155278] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022] Open
Abstract
The discovery of glioblastoma stem cells (GSCs) in the 2000s revolutionized the cancer research field, raising new questions regarding the putative cell(s) of origin of this tumor type, and partly explaining the highly heterogeneous nature of glioblastoma (GBM). Increasing evidence has suggested that GSCs play critical roles in tumor initiation, progression, and resistance to conventional therapies. The remarkable oncogenic features of GSCs have generated significant interest in better defining and characterizing these cells and determining novel pathways driving GBM that could constitute attractive key therapeutic targets. While exciting breakthroughs have been achieved in the field, the characterization of GSCs is a challenge and the cell of origin of GBM remains controversial. For example, the use of several cell-surface molecular markers to identify and isolate GSCs has been a challenge. It is now widely accepted that none of these markers is, per se, sufficiently robust to distinguish GSCs from normal stem cells. Finding new strategies that are able to more efficiently and specifically target these niches could also prove invaluable against this devastating and therapy-insensitive tumor. In this review paper, we summarize the most relevant findings and discuss emerging concepts and open questions in the field of GSCs, some of which are, to some extent, pertinent to other cancer stem cells.
Collapse
Affiliation(s)
- Joana Vieira de Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; (J.V.d.C.); (C.S.G.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Céline S. Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; (J.V.d.C.); (C.S.G.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Adília Hormigo
- Department of Neurology, Neurosurgery, Medicine, The Tisch Cancer Institute and Icahn School of Medicine at Mount Sinai, NY 10029-6574, USA;
| | - Bruno M. Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; (J.V.d.C.); (C.S.G.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
- Correspondence: ; Tel.: +35-1-253-604-872
| |
Collapse
|
18
|
Zhao Z, Bo Z, Gong W, Guo Y. Inhibitor of Differentiation 1 (Id1) in Cancer and Cancer Therapy. Int J Med Sci 2020; 17:995-1005. [PMID: 32410828 PMCID: PMC7211148 DOI: 10.7150/ijms.42805] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Abstract
The inhibitor of DNA binding (Id) proteins are regulators of cell cycle and cell differentiation. Of all Id family proteins, Id1 is mostly linked to tumorigenesis, cellular senescence as well as cell proliferation and survival. Id1 is a stem cell-like gene more than a classical oncogene. Id1 is overexpressed in numerous types of cancers and exerts its promotion effect to these tumors through different pathways. Briefly, Id1 was found significantly correlated with EMT-related proteins, K-Ras signaling, EGFR signaling, BMP signaling, PI3K/Akt signaling, WNT and SHH signaling, c-Myc signaling, STAT3 signaling, RK1/2 MAPK/Egr1 pathway and TGF-β pathway, etc. Id1 has potent effect on facilitating tumorous angiogenesis and metastasis. Moreover, high expression of Id1 plays a facilitating role in the development of drug resistance, including chemoresistance, radiation resistance and resistance to drugs targeting angiogenesis. However, controversial results were also obtained. Overall, Id1 represent a promising target of anti-tumor therapeutics based on its potent promotion effect to cancer. Numerous drugs were found exerting their anti-tumor function through Id1-related signaling pathways, such as fucoidan, berberine, tetramethylpyrazine, crizotinib, cannabidiol and vinblastine.
Collapse
Affiliation(s)
- Zhengxiao Zhao
- Department of Oncology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, China
| | - Zhiyuan Bo
- The Second Department of Biliary Tract Surgery, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai 200438, China
| | - Weiyi Gong
- The Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai 200040, PR China
| | - Yong Guo
- Department of Oncology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, China
| |
Collapse
|
19
|
Whole genome re-sequencing of crested traits and expression analysis of key candidate genes in duck. Gene 2019; 729:144282. [PMID: 31838250 DOI: 10.1016/j.gene.2019.144282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 02/01/2023]
Abstract
The crested duck was a duck breed which features a topknot of feathers on the back of their head. In order to explain the reason of crest, we anatomy the head of some crested ducks. The anatomical structures showed that there was a fat body in the head and a hole in the skull. To determine the reason for the formation of the crest, we used whole genome re-sequencing to detect SNPs and InDels in three crested duck and three normal crested duck (without crest). There were 785,202 unique SNPs and 105,596 unique InDels include in crested duck. There were 14,591 SNPs containing genes and 13,784 InDels continuing genes were mapped on BGI_duck_1.0 by BWA 0.7.16a software. We use KEGG and GO to classification the SNP and InDel containing genes function. The PPI network of SNP containing genes and InDels containing genes was constructed by STRING. The result of PPI and KEGG analysis shown that the formation of crest might include feather development, fatty acid deposition, and skull hypoplasia. To determine the regulated of SNP containing genes and InDels containing genes, which related the different trait, of miRNA we used mirmap to predicted target miRNA of those genes. The miRNA-genes network constructed by Cytoscape. In conclusion, the formation of the crest was a complex process. The fatty acid metabolism block, feather growth and skull hypoplasia might lead crest formation. The tissue expression of four candidate genes showed that they were closely related to the formation of the trait, and could be used as important candidate genes to further elaborate the molecular mechanism of their function.
Collapse
|
20
|
Huang S, Wang LL, Xue NN, Li C, Guo HH, Ren TK, Zhan Y, Li WB, Zhang J, Chen XG, Han YX, Zhang JL, Jiang JD. Chlorogenic acid effectively treats cancers through induction of cancer cell differentiation. Theranostics 2019; 9:6745-6763. [PMID: 31660066 PMCID: PMC6815948 DOI: 10.7150/thno.34674] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/31/2019] [Indexed: 02/06/2023] Open
Abstract
Rationale: Inducing cancer differentiation is a promising approach to treat cancer. Here, we identified chlorogenic acid (CA), a potential differentiation inducer, for cancer therapy, and elucidated the molecular mechanisms underlying its differentiation-inducing effects on cancer cells. Methods: Cancer cell differentiation was investigated by measuring malignant behavior, including growth rate, invasion/migration, morphological change, maturation, and ATP production. Gene expression was analyzed by microarray analysis, qRT-PCR, and protein measurement, and molecular biology techniques were employed for mechanistic studies. LC/MS analysis was the method of choice for chemical detection. Finally, the anticancer effect of CA was evaluated both in vitro and in vivo. Results: Cancer cells treated with CA showed reduced proliferation rate, migration/invasion ability, and mitochondrial ATP production. Treating cancer cells with CA resulted in elevated SUMO1 expression through acting on its 3'UTR and stabilizing the mRNA. The increased SUMO1 caused c-Myc sumoylation, miR-17 family downregulation, and p21 upregulation leading to G0/G1 arrest and maturation phenotype. CA altered the expression of differentiation-related genes in cancer cells but not in normal cells. It inhibited hepatoma and lung cancer growth in tumor-bearing mice and prevented new tumor development in naïve mice. In glioma cells, CA increased expression of specific differentiation biomarkers Tuj1 and GFAP inducing differentiation and reducing sphere formation. The therapeutic efficacy of CA in glioma cells was comparable to that of temozolomide. CA was detectable both in the blood and brain when administered intraperitoneally in animals. Most importantly, CA was safe even at very high doses. Conclusion: CA might be a safe and effective differentiation-inducer for cancer therapy. “Educating” cancer cells to differentiate, rather than killing them, could be a novel therapeutic strategy for cancer.
Collapse
|
21
|
Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor in children with contrasting outcomes. Precise risk assessment contributes to prognosis prediction, which is critical for treatment strategy decisions. In this study, we developed a 3-protein predictor model, including the neural stem cell marker Msi1, neural differentiation marker ID1, and proliferation marker proliferating cell nuclear antigen (PCNA), to improve clinical risk assessment of patients with NB. Kaplan-Meier analysis in the microarray data (GSE16476) revealed that low expression of ID1 and high expression of Msi1 and PCNA were associated with poor prognosis in NB patients. Combined application of these 3 markers to constitute a signature further stratified NB patients into different risk subgroups can help obtain more accurate prediction performance. Survival prognostic power of age and Msi1_ID1_PCNA signature by receiver operating characteristics analysis showed that this signature predicted more effectively and sensitively compared with classic risk stratification system, compensating for the deficiency of the prediction function of the age. Furthermore, we validated the expressions of these 3 proteins in neuroblastic tumor spectrum tissues by immunohistochemistry revealed that Msi1 and PCNA exhibited increased expression in NB compared with intermedial ganglioneuroblastoma and benign ganglioneuroma, whereas ID1 levels were reduced in NB. In conclusion, we established a robust risk assessment predictor model based on simple immunohistochemistry for therapeutic decisions of NB patients.
Collapse
|
22
|
Kim TJ, Kwon HS, Kang M, Leem HH, Lee KH, Kim DY. The Antitumor Natural Compound Falcarindiol Disrupts Neural Stem Cell Homeostasis by Suppressing Notch Pathway. Int J Mol Sci 2018; 19:ijms19113432. [PMID: 30388862 PMCID: PMC6274977 DOI: 10.3390/ijms19113432] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 10/25/2018] [Accepted: 10/31/2018] [Indexed: 01/01/2023] Open
Abstract
Neural stem cells (NSCs) are undifferentiated, multi-potent cells that can give rise to functional neurons and glial cells. The disruption in NSC homeostasis and/or the impaired neurogenesis lead to diverse neurological diseases, including depression, dementia, and neurodegenerative disorders. Falcarindiol (FAD) is a polyacetylene found in many plants, and FAD shows the cytotoxicity against breast cancers and colon cancers. However, there is no research on the consequence of FAD treatment in normal stem cells. Here, we suggest that FAD has anticancer roles against glioblastoma cells by inducing the differentiation of glioblastoma stem-like cells, as well as activating apoptosis pathway in glioblastoma cells. On the other hand, we also show that FAD has detrimental effects by disrupting the maintenance of normal NSCs and altering the balance between self-renewal and differentiation of NSCs.
Collapse
Affiliation(s)
- Tae-Jun Kim
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu 41940, Korea.
| | - Hyun-Sook Kwon
- National Development Institute of Korean Medicine, Gyeongsan, Gyeongsangbuk-do 38540, Korea.
| | - Mingyu Kang
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu 41940, Korea.
| | - Hyun Hee Leem
- National Development Institute of Korean Medicine, Gyeongsan, Gyeongsangbuk-do 38540, Korea.
| | - Kyung-Ha Lee
- Department of Cosmetic Science and Technology, College of Bio-industry, Daegu Haany University, Gyeongsan 38610, Korea.
| | - Do-Yeon Kim
- Department of Pharmacology, School of Dentistry, Brain Science and Engineering Institute, Kyungpook National University, Daegu 41940, Korea.
| |
Collapse
|
23
|
Testa U, Castelli G, Pelosi E. Genetic Abnormalities, Clonal Evolution, and Cancer Stem Cells of Brain Tumors. Med Sci (Basel) 2018; 6:E85. [PMID: 30279357 PMCID: PMC6313628 DOI: 10.3390/medsci6040085] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/19/2018] [Accepted: 09/25/2018] [Indexed: 02/06/2023] Open
Abstract
Brain tumors are highly heterogeneous and have been classified by the World Health Organization in various histological and molecular subtypes. Gliomas have been classified as ranging from low-grade astrocytomas and oligodendrogliomas to high-grade astrocytomas or glioblastomas. These tumors are characterized by a peculiar pattern of genetic alterations. Pediatric high-grade gliomas are histologically indistinguishable from adult glioblastomas, but they are considered distinct from adult glioblastomas because they possess a different spectrum of driver mutations (genes encoding histones H3.3 and H3.1). Medulloblastomas, the most frequent pediatric brain tumors, are considered to be of embryonic derivation and are currently subdivided into distinct subgroups depending on histological features and genetic profiling. There is emerging evidence that brain tumors are maintained by a special neural or glial stem cell-like population that self-renews and gives rise to differentiated progeny. In many instances, the prognosis of the majority of brain tumors remains negative and there is hope that the new acquisition of information on the molecular and cellular bases of these tumors will be translated in the development of new, more active treatments.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, 00161 Rome, Italy.
| |
Collapse
|
24
|
Autophagy in glioma cells: An identity crisis with a clinical perspective. Cancer Lett 2018; 428:139-146. [PMID: 29709703 DOI: 10.1016/j.canlet.2018.04.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/14/2018] [Accepted: 04/20/2018] [Indexed: 01/06/2023]
Abstract
Over the last decade, autophagy has emerged as one of the critical cellular systems that control homeostasis. Besides management of normal homeostatic processes, autophagy can also be induced by tissue damage stress or by rapidly progressing tumors. During tumor progression, autophagy mediates a cellular reaction to the changes inside and outside of cells, which leads to tumor adaptation. Even though the regulation of autophagy seems universal and is a well-described process, its dysregulation and role in glioma progression remain an important topic of investigation. In this review, we summarize recent evidence of autophagy regulation in brain tumor tissues and possible interconnection between signaling pathways that govern cellular responses. This perspective may help to assess the qualitative differences and various outcomes in response to autophagy stimulation.
Collapse
|
25
|
Huang H, Wang C, Liu F, Li HZ, Peng G, Gao X, Dong KQ, Wang HR, Kong DP, Qu M, Dai LH, Wang KJ, Zhou Z, Yang J, Yang ZY, Cheng YQ, Tian QQ, Liu D, Xu CL, Xu DF, Cui XG, Sun YH. Reciprocal Network between Cancer Stem-Like Cells and Macrophages Facilitates the Progression and Androgen Deprivation Therapy Resistance of Prostate Cancer. Clin Cancer Res 2018; 24:4612-4626. [PMID: 29691294 DOI: 10.1158/1078-0432.ccr-18-0461] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/03/2018] [Accepted: 04/20/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Cancer stem-like cells (CSC) contribute to the progression and androgen deprivation therapy (ADT) resistance of prostate cancer. As CSCs depend on their specific niche, including tumor-associated macrophages (TAM), elucidating the network between CSCs and TAMs may help to effectively inhibit the progression and ADT resistance of prostate cancer.Experimental Design: The underlying intracellular mechanism that sustains the stem-like characteristics of CSCs in prostate cancer was assessed via RNA sequencing, co-immunoprecipitation, chromatin immunoprecipitation, and other assays. A coculture system and cytokine antibody arrays were used to examine the interaction network between CSCs and TAMs. In addition, an orthotopic prostate cancer model was established to evaluate the in vivo effects of the combined targeting of CSCs and their interaction with TAMs on ADT resistance.Results: Autophagy-related gene 7 (ATG7) facilitated the transcription of OCT4 via β-catenin, which binds to the OCT4 promoter, promoting CSC characteristics in prostate cancer, including self-renewal, tumor initiation, and drug resistance. In addition, CSCs remodeled their specific niche by educating monocytes/macrophages toward TAMs, and the CSC-educated TAMs reciprocally promoted the stem-like properties of CSCs, progression and ADT resistance of prostate cancer via IL6/STAT3. Furthermore, the combined targeting of CSCs and their interaction with TAMs by inhibiting ATG7/OCT4 and IL6 receptor effectively ameliorated ADT resistance in an orthotopic prostate cancer model.Conclusions: Targeting CSCs and their niche may prove to be a more powerful strategy than targeting CSCs alone, providing a rational approach to ameliorating ADT resistance in prostate cancer. Clin Cancer Res; 24(18); 4612-26. ©2018 AACR.
Collapse
Affiliation(s)
- Hai Huang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Chao Wang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Fei Liu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hui-Zhen Li
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Guang Peng
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xu Gao
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Ke-Qin Dong
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hong-Ru Wang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - De-Pei Kong
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Min Qu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Li-He Dai
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Kai-Jian Wang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhe Zhou
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jun Yang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Ze-Yu Yang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yan-Qiong Cheng
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Qin-Qin Tian
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Dan Liu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Chuan-Liang Xu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Dan-Feng Xu
- Department of Urinary Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xin-Gang Cui
- Department of Urinary Surgery, The Third Affiliated Hospital of Second Military Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, China.
- Department of Urinary Surgery, Gongli Hospital, Second Military Medical University, Shanghai, China
| | - Ying-Hao Sun
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China.
| |
Collapse
|
26
|
Kim TJ, Byun JS, Kwon HS, Kim DY. Cellular toxicity driven by high-dose vitamin C on normal and cancer stem cells. Biochem Biophys Res Commun 2018; 497:347-353. [DOI: 10.1016/j.bbrc.2018.02.083] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 02/08/2018] [Indexed: 12/20/2022]
|